Open access peer-reviewed chapter - ONLINE FIRST

Trends of Diagnostic Methods for Human Viral Diseases

Written By

Biswajyoti Borkakoty, Aniruddha Jakharia, Pratibha Singh and Siraj Ahmed Khan

Submitted: 02 August 2023 Reviewed: 18 September 2023 Published: 06 March 2024

DOI: 10.5772/intechopen.113239

Viral Infectious Diseases - Annual Volume 2024 IntechOpen
Viral Infectious Diseases - Annual Volume 2024 Authored by Shailendra K. Saxena

From the Annual Volume

Viral Infectious Diseases - Annual Volume 2024 [Working Title]

Prof. Shailendra K. Saxena

Chapter metrics overview

55 Chapter Downloads

View Full Metrics

Abstract

The global health field is significantly affected by viral infections, and sero-diagnosis is crucial in diagnostic virology. Various laboratory techniques such as nucleic acid detection, viral culture, and antigen detection are essential for diagnosing viral infections. Advances in science have led to the development of new immunologic and molecular techniques, enabling rapid and simplified diagnosis of different viruses. Timely and accurate identification of viral infections is vital for effective outbreak management. Immunological techniques, detecting viral antigens or antibodies, are widely used in diagnostic and epidemiological research, aiding in epidemic identification, appropriate diagnostic tests, vaccination programs, and detecting common and emerging viruses. However, traditional viral identification methods demand extensive technical expertise, time, and financial resources. Consequently, scientists worldwide are dedicated to developing precise diagnostic methods for viral diseases. Various innovative approaches are being explored, aiming to create more accessible, time-efficient, and cost-effective viral disease diagnosis methods, thereby benefiting low-income countries.

Keywords

  • viral infection diagnosis
  • viral serology
  • real-time PCR
  • next-generation sequencing
  • NGS
  • RPA
  • LFA
  • CRISPR-Cas 9

1. Introduction

The Baltimore classification system and the International Committee on Taxonomy of Virus system both categorized viruses primarily on a small number of morphological characteristics. The Baltimore categorization system is based on two factors: the kind of genome (such as RNA or DNA, whether it is double- or single-stranded, etc.) and the mechanism of replication. Viral infection impacts millions of people every year and contributes to illness and death from serious, chronic, recurrent, or life-threatening disorders. The development of complicated and accurate mechanisms for detection of nucleic acid of virus and the antiviral antibody has potential to increase accuracy at various stages of the disease. Advancing diagnostic procedures in the past decades has significantly reduced the time and improved the accuracy of viral diagnosis, helping with patient care, disease control, and favorable impact on disease outcomes. Detection of viral antigen and antibody and viral nucleic acids are some of the more common methods for determining the presence of pathogens in humans. Most recently, the Real-Time PCR (qPCR) method has been widely employed in viral diagnostics due to its highly specific DNA/RNA detection and high-throughput quantification. Antiviral susceptibility testing, viral genotyping, and on-site care have all been developed as a result of rapid diagnosis leading to better and early treatment support. Laboratory-controlled serological tests have the benefits of high specificity, sensitivity, and differential diagnostic choices. Advancement in molecular and serological techniques has made it easier to diagnose viral infections [1, 2, 3, 4].

Every year, viral diseases claim the lives of millions of people worldwide, contributing to a significant global mortality rate. Due to seasonal influenza, there are billions of cases annually with 3–5 million cases of severe illness, leading to 290,000 to 650,000 respiratory deaths annually [5]. The swine influenza A (H1N1) in 2009 was declared a pandemic by WHO as it caused an estimated 284,400 deaths worldwide during the year. In 2010, rabies caused an estimated 61,000 deaths, with a substantial 84% of these occurring in rural areas. It is also estimated that 59,000 people die each year due to rabies [6]. During 2019 1.1 million people died due to viral hepatitis and it is also estimated that 354 million people live with hepatitis B or C worldwide. Acute hepatitis and chronic infection lead to liver cancer and cirrhosis and globally millions of people lose their live yearly [7]. More recently, the world faced unprecedented challenges in the form of the COVID-19 pandemic, triggered by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) [2]. World Health Organization (WHO) reported an alarming 771,407,825 confirmed cases of COVID-19 with 6,972,152 deaths till September 2023 (https://covid19.who.int/). In the face of these alarming statistics, the precise and early detection of viral infections emerges as a crucial aspect for effective disease management. Early detection not only enables prompt treatment but also plays a pivotal role in reducing treatment costs, lowering mortality rates, and gaining control over the spread of the disease. Furthermore, diagnostic methods such as variant identification and characterization facilitate early identification of viral variants, development of new anti-viral agents, and antimicrobial vaccines, which will augment the implementation of effective preventative measures. Conventionally, diagnostic methods relied on serological approaches, which revolutionized the diagnosis of viral infections in both humans and animals. However, these conventional methods have proven to be less sensitive, time-consuming, and difficult to perform in clinical samples other than blood. To overcome the limitations of conventional procedures, there is an urgent need for fresh and innovative approaches. Molecular methods, particularly those aimed at detecting viral nucleic acids, have emerged as promising solutions. These advanced techniques offer higher sensitivity and specificity, revolutionizing the diagnosis of viral infections. Embracing cutting-edge technology allows us to enhance the accuracy and efficiency of viral detection, ultimately leading to improved patient outcomes and effective control of viral outbreaks. Implementation of innovative approaches in viral diagnostics will serve as a significant step toward safeguarding the well-being of communities worldwide [5].

Advertisement

2. Diagnostic techniques for viruses

2.1 Evolution of diagnostic tools

There have been many developments in diagnostic techniques (molecular and serological) for viral antigens or antibodies in the past many years. Diagnostic techniques have evolved through the ages and are still evolving (Table 1).

YearDiagnostic techniquesYearDiagnostic technique
1930Complement fixation1985–1999ELISA, western blot, immunostaining
1948Cell culture1986Fluorescence in situ hybridization (FISH)
1949Hemagglutination inhibition1989Nucleic acid sequence-based amplification
1954Electron microscopy1990Immunostaining
1960Nanotechnology concept1991Chemiluminescent immunoassay
1964Immunofluorescence assay1992Concept of real-time PCR
1966Particle agglutination1995Gene chip
1968Radioimmunoassay1996DNA microarray
1970Development of nucleic acid hybridization1996Pyrosequencing technique—The next-generation Sequencing
1973Enzyme immunoassay1999Nanotechnology and virus detection
1975Southern blotting technique2000Loop-mediated isothermal amplification
1977Sanger sequencing2005454 Pyrosequencer system
1978Dot hybridization2005qRT-PCR, virus microarrays
1980Polymerase chain reaction2006Illumina/Solexa
1980Maxim Gilbert Sequencing method2007ABI/SoLID sequencing
1985Restriction fragment length polymorphism analysis (RFLP)2013CRISPR system
1985Immunoblotting assay2015VirCapSeq-VERT

Table 1.

Timeline for the development of various serological and molecular detection techniques constituting the mainstream approaches of both laboratory research and clinical diagnosis of viruses [8, 9].

2.2 Traditional method for virus identification

Isolating the virus through tissue culture, animal models, or embryonated chicken eggs was the traditional laboratory method for isolation and identification of medically important viruses [10].

2.2.1 Virus culture

Virus culture is the “gold standard” method for diagnosing culturable viruses. Viruses are cultivated in cell culture (Table 2) and cells infected with viruses exhibit morphological alterations that vary according to the virus’s properties (Figure 1). This technique is important, since it is the only approach that produces viable isolates for further characterization. However, it requires well-equipped bio-safety level laboratories and is time-consuming [10, 12, 13, 14].

Virus familyVirusCell line
CaliciviridaeNorovirus (NV)HG23
Murine norovirus-1 (MNV-1)RAW267.4
CoronaviridaeHuman coronavirus 229E (229E)MRC-5
SARS-CoV-2Vero
PicornaviridaeHepatitis A virus (HAV)FRhK-4
Enterovirus 71 (EV71)Vero
Human rhinovirus strains 18, 51, and 68 (HRV 18, 51, and 68)HeLa
AdenoviridaeAdenovirus 1Human embryo kidney cell
HepadnaviridaeHepatitis B virusHepaRG

Table 2.

Viruses and respective culturable cell lines [11].

Figure 1.

(A). MDCK cell line culture (B). Infection with respiratory sample (C). Hemagglutination inhibition assay (D). Serial dilution method for virus culture.

2.2.2 Microscopy

Microscopy is considered a classic method for diagnosis/identification of viruses through direct visualization of viruses. Different types of microscopy are as follows:

2.2.2.1 Light microscopy

The light microscope, a vital tool in biology, identifies and magnifies objects by transmitting light through lenses. Replicating viruses induce distinctive histological changes in infected cells, often forming inclusion bodies, such as Negri bodies in rabies and cytomegalic inclusion bodies in CMV infections. These changes serve as diagnostic indicators in virology [15].

2.2.2.2 Electron microscope

Electron microscopy (EM) is a technique that allows direct visualization of viruses in clinical specimens and viral isolates. It is an essential tool in the detection, analysis of virus replication, and investigation of virus-host cell interactions. Identification by EM is based on morphological features of the virus, such as size, shape, and ultra-structural features. The constraint of electron microscopy is that sufficient viruses must be present (approx. 105–106 particles/mL). EM is not used widely for routine diagnosis because it is expensive and gives black-and-white images, and only live specimens can be visualized [6].

2.3 Serological/immunological diagnostic techniques

The natural immune response to viral infection has served as the foundation for the development of serological/immunological diagnostic methods. Antigen-antibody complex formation is the foundation for serological assays for the detection of whole viruses or viral antigens/viral antibodies, etc. Several assay procedures were developed for viral antigen or antibody detection:

2.3.1 Enzyme immunoassay

The term “enzyme immunoassay” is also referred to as the enzyme-linked immunosorbent assay (ELISA). It is more sensitive and capable of detecting ≤1 nanogram of viral antigen or antibodies per ml in the clinical specimen. The basic concept of this technique is to identify/detect the antigen or antibody of interest from the rest of the component present in the assay specimen/sample (Figure 2) [16].

Figure 2.

Schematic representation of enzyme immunoassay.

There are four major types of ELISA [17]:

  • Direct ELISA- In a direct ELISA, the antigen (sample/analyte) is immobilized on the surface of the multi-well plate and blocking is done with suitable blocking agents like BSA to avoid non-specific binding. Sample is added followed by addition of enzyme-conjugated-specific antibody which directly binds to the antigen of interest. Upon addition of substrate, there is a change of color which helps in the detection of antigen of interest in the sample. Proper washing steps are involved between each step except after the addition of substrate.

  • Indirect ELISA- An indirect ELISA is similar to direct ELISA, specific antigen is immobilized on the surface of the multi-well plate, and blocking is done similarly as in direct ELISA. The sample/analyte (primary antibody) is added, which binds to the immobilized antigen. The secondary antibody (anti-immunoglobulin antibody) conjugated with enzyme is added, which binds to the primary antibody. Similarly, direct ELISA substrate addition is done for detection.

  • Sandwich ELISA- It is the most commonly used ELISA test for diagnostic purposes. It requires two antibodies specific for different epitopes of the antigen. These two antibodies are normally referred to as matched antibody pairs. Capture antibody is immobilized on the surface of the multi-well plate, and blocking is done. The sample/analyte is added, which binds to the immobilized antibody. Followed by the addition of detection antibody conjugated with enzyme. Similarly, direct ELISA substrate addition is done for detection.

  • Competitive ELISA-Competitive ELISAs are commonly used for small molecules when the protein of interest is too small to efficiently sandwich with two antibodies. It is used to test for the presence of an antibody specific for antigens in the test serum. This type of ELISA utilizes two specific antibodies, an enzyme-conjugated antibody and another antibody present in the test serum (if the serum is positive). Combining the two antibodies into the wells will allow for competition for binding to antigens. The presence of a color change means that the test is negative because the enzyme-conjugated antibody bound the antigens (not the antibodies of the test serum). The absence of color indicates a positive test and the presence of antibodies in the test serum. The competitive ELISA has a low specificity and cannot be used in dilute samples. However, the benefits are as follows: It can measure a large range of antigens in a given sample, it can be used for small antigens, and it has low variability. Example—West Nile virus, Hepatitis A virus antibody, and Hepatitis B virus antibody.

2.3.2 Radioimmunoassay and time-resolved Fluoroimmunoassay

Enzyme immunoassay (EIA) precedes radioimmunoassay (RIA). The sole difference is that the label is a radioactive isotope like 125I rather than an enzyme and a gamma (γ) counter is used to quantify bound antibodies. A known quantity of antibodies is combined with radiolabeled antigens that specifically bind to each other. An undetermined amount of the same antigen is introduced to a patient’s serum sample. Due to this, the radiolabeled antigen and the unlabeled antigen from the serum compete for binding sites on antibodies. When there is an increase in the amount of unlabeled antigen, there is more binding to its antibody, as a result radiolabeled antigen is replaced and overall, there is a decrease in the ratio of antibody and bound radiolabeled antigen to free radiolabeled antigen. Following the separation of the bound antigens, the radioactivity of the free (unbound) antigen that is still present in the supernatant is determined using a gamma counter. Although this is a reliable and sensitive assay, it is costly, and the radioactive material is toxic. Time-Resolved Fluoroimmunoassay (TR-FIA) is a non-isotopic immunoassay, which uses fluorophore to tag indicator antibodies. The fluorophore generates fluorescence at a distinct wavelength when excited by light, which can be quantified using a time-lapse fluorometer. The target antibody or antigen is marked using the time-resolved fluorescence immunoassay (TR-FIA) technique, which makes use of lanthanide elements with distinctive fluorescence properties and the associated chelates as markers [18, 19]. It frequently makes use of antibody that is adhered to the microplate well’s bottom. The antibody binds to the target molecule on the plate, while the samples are incubated in these wells. After washing the plate to remove any unbound samples, a secondary antibody that is covalently attached to the most commonly used lanthanide chelate, europium, is added, while the secondary antibody that is not attached is washed away. The amount of lanthanide-labeled antibody present is directly correlated with the concentration of the target molecule present in the sample. The auto-fluorescence signal’s decay serves as the time gate for the detection. This indicates that the short-lived auto-fluorescence signal must have completely faded before time-resolved fluorescence detection may begin. Integrated intensity is used to measure the data instead of time decay, and the detected emission signal is integrated across a certain time range. A standard curve can be used to quantify the analyte amount since it is proportional to the time-resolved emission signal. It has the same sensitivity as RIA but requires more expensive equipment [20]. Examples: Varicella Zoster virus and Lyssa virus can be diagnosed by this method (Figure 3).

Figure 3.

Diagrammatic representation of (A) radioimmunoassay and (B) time-resolved fluoroimmunoassay.

2.3.3 Chemiluminescence immunoassay

It is the technique in which the label/conjugate used is the luminescent molecule. Luminescence is the emission of visible or near-visible radiation (300–800 nm), which is generated when an electron transitions from an excited state to a ground state and energy in the atom gets released in the form of light which is measured in relative light unit. Chemiluminescent can be direct using luminophore (acridinium and ruthenium esters) markers or indirect using enzyme markers (alkaline phosphatase with adamantyl 1,2-dioxetane aryl phosphate and horseradish peroxidize with luminal or its derivative as substrate). Antigen or antibodies are directly labeled with chemiluminescent agent and emit light after oxidizing and emission of light is measured. It is more sensitive than ELISA and has a long luminous time, and it takes less time for analysis of sample [21].

2.3.4 Latex particle agglutination

It is the simplest immunoassay available. It is accomplished through the agglutination of tiny latex beads coated with antiviral antibodies with antigen. Collected sample were mixed with latex beads coated with specific antigen or antibody in normal saline. If antigen or antibody of the virus of interest is present, the beads will agglutinate together and the result can be viewed in minutes. However, due to limited sensitivity and specificity, false-negative results are common in samples with insufficient number of virions. Examples: avian influenza virus and Lyssa virus can be diagnosed by this method (Figure 4) [22].

Figure 4.

Diagrammatic representation of (A) positive agglutination test for antibody. (B) Positive agglutination test for antigen.

2.3.5 Immunochromatography

Immunochromatography is used to determine the movement of antigen or complexes of antigen and antibody via support material such as agarose gel, filter paper, or nitrocellulose membrane. A standard format involves tagged antibody to react with an unidentified antigen, and the antigen-antibody complex migrates through the solid support via capillary action, allowing the complex to be detected if antigens are present. Positive control and negative control are used to confirm the validity of individual tests [22]. Examples: Adenovirus [23], Rotavirus, and Norovirus can be diagnosed using this method (Figure 5) [24].

Figure 5.

Diagrammatic representation of immunochromatography. (A) Control test. (B) Positive immunochromatographic test. (C) Negative immunochromatographic test.

2.3.6 Viral cytopathology (cytology/histology)

Rapid diagnosis began with the investigation of viral cytopathology. Herpes virus infections diagnosed by using Tzank preparation are an example of this type of testing. The skin vesicle base is scraped and transferred to a microscope slide. After the slide is dried naturally, it is stained with Wright, Giemsa, or Papanicolaou stain. A normal microscope is used to view the slides. For viruses such as cytomegalovirus (CMV), Varicella zoster virus (VZV), Human Papillomavirus (HPV), BK virus, and parovirus B19, and particular cytological alterations can be validated by staining for specific antigens, antibodies, or nucleic acid probes. It includes immunohistochemistry and in situ hybridization [6].

2.3.6.1 Detection of antigen in tissues (immunohistochemistry)

Antigens from viruses can be detected in sections of frozen tissue or exfoliated cells when tissue or cells are fixed on a glass slide, which serves as a solid support. When activated by shorter-wavelength lights, an antigen and antibody combination that has been tagged by a fluorochrome produces light of a specified long wavelength. In a normal microscope, this light can be seen as fluorescence after other wavelengths have been filtered. However, the sensitivity of this approach is insufficient for detecting fluorescent antibody complexes with soluble antigens or virions. The approach is classified into two distinct subtypes. They are as follows: direct immunofluorescence and indirect immunofluorescence [2, 6, 22]. Examples- Human herpesviruses, Epstein-Barr virus, and cytomegalovirus can be diagnosed by this method (Figure 6) [25].

Figure 6.

Diagrammatic representation of immunohistochemistry. (A) Indirect immunofluorescence. (B) Direct fluorescence.

2.3.7 Complement fixation

In the presence of RBC, the antigen-antibody complex binds complement, rendering it critical for the lysis of RBC. It consists of two stages. The first stage involves mixing guinea pig serum, which includes the complement, with a specific antigen and test serum. The second stage of the response cannot occur if particular antibodies are present because the complement joins with the antigen-antibody complex. The second stage involves adding erythrocytes from sheep that were previously exposed to rabbit serum anti-sheep erythrocytes (hemolytic system). No hemolysis will take place if the complement has been fixed in the first step (positive reaction). On the other hand, the complement, which does not bind, can interact with the hemolytic system and lyse sheep erythrocytes if the blood test does not include appropriate antibodies (a negative reaction) [26]. Examples: Hepatitis virus and Lyssa virus can be diagnosed by this method (Figure 7) [27].

Figure 7.

Diagrammatic representation of (A). Complement fixation (B). No complement fixation.

2.3.8 Immunoperoxidase staining

Similar to IF, immune peroxidase (IP) methods use a conjugate that is an enzyme, like horseradish peroxidase. While an anti-animal species antibody is labeled in the indirect technique, the virus-specific antibody is enzyme-tagged in the direct method. Addition of substrate, such as aminoethyl carbazole or di-amino benzidine, and oxidizing in the presence of hydrogen peroxide result in a reddish-brown color, and the virus-infected cells bound to enzyme conjugate can be observed. IP staining has a number of advantages over IF, although it requires an additional step (the addition of the substrate). With the naked eye or a light microscope, the reaction can be observed. The reagents are more stable, the slides are more permanent, and there are fewer nonspecific reactions [28]. Although endogenous peroxidase, which is found in a variety of tissues, most notably leukocytes, might give a false positive, this issue can be avoided with good procedures and enough controls [22]. Examples: Baculovirus can be diagnosed by this method (Figure 8) [29].

Figure 8.

Diagrammatic representation of immunoperoxidase staining.

2.3.9 Hemagglutination inhibition assay (HIA)

Certain viruses carry an antigen called hemagglutinin on their surface that attaches and agglutinates RBCs. This process is referred to as hemagglutination (HA). The ability of viruses to agglutinate RBCs was used to develop the HI technique. On a microtiter plate, serial dilutions of serum samples are made for the HI test. Following that, a predetermined number of viral hemagglutinins and suitable RBCs were added. In the absence of hemagglutinins, favorable reactions are seen. This is determined by microtiter plate tilting, which allows for the streaming of free RBCs (Figure 9) [2].

Figure 9.

Diagrammatic representation of Hemagglutination inhibition assay. (A). Control assay. (B). Hemagglutination assay. (C). Hemagglutination inhibition assay.

2.3.10 Western blotting analysis

This technique, alternatively called immunoblotting, is used for detecting antiviral antibodies or the viral protein by denaturing and separating entire viral protein on Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE). Then, viral proteins are electro-transferred to nitrocellulose membranes. After that, the membrane is treated with antibodies linked to enzymes specific for the viral proteins. When enzyme-labeled antibodies bind to viral proteins, chromogenic substrate is added, which results in colorful bands at the locations of the viral antigens [2, 22]. Examples: Lyme disease virus, human immunodeficiency virus (human immunodeficiency), and Hepatitis B virus can be diagnosed by this method (Figure 10).

Figure 10.

Diagrammatic representation of Western blotting assay.

2.3.11 Neutralization assays

Neutralization assays for viruses are much specialized assays that neutralize antibodies. They are also employed to verify the results of other types of serological assays, like ELISA, that have been shown to cross-react with viruses belonging to the same family, for example, the Flaviviruses (Dengue virus, West Nile virus, and Zika virus). Antigen is combined with antibody dilutions, and the prevention of cytopathic effects (CPE) is observed via inoculation on a monolayer of tissue. The inhibitory impact can be determined either by observing the CPE or by overlaying agar that allows for plaque formation in plaque reduction neutralization test (PRNT) [30].

2.3.11.1 Plaque reduction neutralization test (PRNT)

The plaque reduction neutralization test is used to determine the titer of antiviral antibodies. A viral suspension is combined with a diluted serum sample or antibody testing solution. For the interaction of antibodies with the virus, the sample is incubated. To stop the virus from spreading carelessly, carboxymethyl cellulose or agar is applied to the surface of the cell layer. It is possible to estimate the concentration of plaque-producing units by counting the number of plaques (regions of infected cells) that appear after a few days. Depending on the virus, fluorescently labeled antibodies or certain dyes reacting with infected cells are used to measure the plaque-forming units. For lowering the number of plaques by 50%, the amount of serum needed when compared to the serum-free virus provides a measurement of the antibody’s level or potency. The PRNT50 value is used to refer to this measurement. Example: Dengue virus can be diagnosed by this method (Figure 11) [30].

Figure 11.

Diagrammatic representation of plaque reduction neutralization test.

2.3.12 miRNA-based Immunodetection

miRNAs, a class of short conserved non-coding RNAs comprising 19–24 nucleotides, play a crucial role in post-transcriptional regulation. It has been recently found that for various malignancies, exogenous miRNA is potential diagnostic marker, including Hepatocellular Carcinoma (HCC), as part of immunotechniques. In HCC, Alpha-fetoprotein, exosomes, and serum microRNAs have shown promise as diagnostic markers. Another noteworthy study introduced a synthetic miRNA-based approach to produce neutralizing antibodies, which are directly expressed in the lungs through aerosol administration. This method aims to combat Respiratory Syncytial Virus (RSV) and influenza infections in humans. Recent research highlights the evolutionary conservation of endogenous miRNAs, and their presence in body fluids indicates their role in regulating pathogenesis, immune responses, and viral infections. These circulating miRNAs have proven valuable for early diagnosis of viral infections, with diagnostic techniques relying on biomarkers. Numerous circulating microRNA biomarkers have been identified for diagnostic purposes, such as miR29b and miR125 for hepatitis infection, miR155 and miR1260 for influenza, and miR12, miRVP3p, and miR184 for arboviruses. These discoveries have significantly enhanced the early detection and management of viral infections [1, 31].

2.3.13 Metagenomics-based immunodetection

Successful testing of the mNGS pan-pathogen detection assay has shown promise in patients with acute viral infections. To efficiently analyze Next-Generation Sequencing (NGS) data, an innovative bioinformatics pipeline called SURPI+ was developed. This pipeline enables the automatic generation of pathogen summaries and visualization of results through a graphical user interface. In a recent breakthrough, metagenomic next-generation sequencing was applied in diagnosing SLEV meningoencephalitis. Notably, studies reveal that metagenomic viral sequencing has the potential to identify viruses beyond influenza. Respiratory sample sequencing utilizing Oxford Nanopore Technology has been explored, offering significant potential for diagnosing various clinical respiratory viruses when compared to current standard diagnostic approaches. The nanopore platform holds great promise for enhancing virus detection in clinical settings [1, 32].

2.3.14 Monoclonal antibodies-based immunodetection

In recent times, the development of therapeutic and aptamer-based diagnostic platform technologies has shown promise in diagnosing viral infections. Oligonucleotide aptamers, in particular, have emerged as prospective alternatives to monoclonal antibodies for detection. They possess the unique ability to target various proteins within infected cells or viral particles, making them innovative and versatile diagnostic agents. In the future, these aptamer molecules hold the potential to serve as viable alternatives to traditional monoclonal antibodies in diagnostic applications [1, 33].

2.3.15 Microfluidic technology-based immunodetection

Microfluidics has enabled significant advancements in the study of viral fusion kinetics, viral infectivity, and the screening of neutralizing molecules for viral responses. Additionally, microfluidic platforms hold great promise as clinical tools for diagnostics and drug screening. The field of microfluidics emphasizes precise control and manipulation of fluids within microscopic channels, supporting various biological research methods, including flow cytometry, DNA analysis, enzyme tests, and immunoassays. Recent research has explored the use of integrated microfluidic systems for diagnostic purposes. For instance, aptamers targeting Influenza A viruses (IAV H1N1) were utilized to detect the virus. The complexity and variability of viral infections, heavily influenced by the chemical and physical environment, make microfluidics an ideal platform for conducting studies on infection biology. This technology offers unique insights and opportunities to advance our understanding of viral infections and develop improved diagnostic and therapeutic approaches (Figure 12) [1].

Figure 12.

Diagrammatic representation of (A) IgG or IgM antibodies, which are detected on microfluidic chip using fluorescence detection. (B) Antigen, which is detected on microfluidic chip using fluorescence detection.

2.3.16 Nanoparticles-based immunodetection

The emerging new class of nanoscale materials, possessing diverse and remarkable features, has sparked a wave of innovation in biological and diagnostic applications. Nanoparticle-based lateral flow immunoassays (LFIAs) are used as a point-of-care diagnostic tool for detecting disorders and infectious viral agents. LIFA is a paper-based point-of-care strip biosensor used to identify specific analytes in virus-infected samples. Detection strategies based on gold nanoparticles (AuNP) have demonstrated effectiveness for various viruses, with special emphasis on utilizing bio-AuNP hybrid structures and employing recently developed assay techniques and formats. Notable examples of viruses, where LFIA and AuNP-based strategies have been applied, include Ebola virus, Dengue virus, Human cytomegalovirus, West Nile virus, Human Papillomavirus, and more. These innovative approaches hold significant promise for rapid and accurate diagnosis in critical healthcare scenarios (Figure 13) [1, 34].

Figure 13.

Diagrammatic representation of nanoparticles-based immunodetection.

2.3.17 Immunosensor-based immunodetection

Immunodetection can be accomplished using an immunosensor. A selective and sensitive electrochemical immunosensor for early-stage diagnostics can be constructed by integrating an immune sensing chip with a miniaturized potentiostat and connecting it to a smartphone. Researchers designed a low-cost, portable graphene-enabled biosensor with an immobilized monoclonal antibody that is highly selective. Field Effect Biosensing (FEB), which depends on covalently attaching monoclonal antibodies to graphene, allows for the real-time quantitative detection of antigens. An electrochemiluminescence (ECL) immunosensor of the sandwich type was developed for the ultrasensitive detection of surface antigens [1]. Examples: Influenza virus can be diagnosed by this method (Figure 14) [35].

Figure 14.

Diagrammatic representation of immunosensor-based immunodetection.

2.3.18 Surface-enhanced Raman spectroscopy (SERS)

Surface-enhanced Raman spectroscopy (SERS) is becoming a vital tool in bioanalytical applications, such as the identification and categorization of pathogenic organisms because it allows for the quick detection of analytes with chemical specificity inherent to vibrational spectroscopy. Driskell et al. [36] used SERS to detect strains of rotavirus by determining their molecular signature. PLS-DA, a chemometric data analysis tool, made it easier to classify viruses on the basis of differences in spectrum. Its application in determining complexities, physical characteristics of molecules is more impressive and highly favorable in comparison of using tagged reporters in an indirect manner with fluorochrome or radioactive materials. The disadvantage is it is labor expensive and time-consuming, depending on the operator’s abilities, so as a result, automation is required. Examples: Influenza A virus subtypes and SARS-COV-2 can be diagnosed by this method (Figure 15).

Figure 15.

Diagrammatic representation of surface-enhanced Raman spectroscopy.

2.4 Molecular/nucleic acid-based diagnostic technique

Nucleic acid-based molecular diagnostic methods have brought about a revolutionary change in diagnostic virology. These techniques offer speed, high sensitivity, and specificity, allowing for the diagnosis of nearly all viruses known to cause human illnesses by identifying specific nucleic acid sequences. They enable rapid detection and can simultaneously identify numerous viruses, including those that are challenging to culture, slow-growing, or possess antigenic variants. The extensive adoption of this technology can be attributed to the combination with other improved techniques. Furthermore, continuous advancements in sequencing technologies for nucleic acids, along with the available sequence data in databases, have greatly facilitated the analysis of collected data. This progress has opened up new possibilities in diagnosing viral infections more accurately and efficiently [22].

2.4.1 Polymerase chain reaction

It is the more sensitive approach for detecting the presence of undetectable genome (DNA or RNA) of viruses. It is a procedure that can be used in place of direct hybridization. It is used for the amplification of a short fragment of viral RNA or DNA found in the clinical specimen up to a millionfold, enabling the detection of infectious agents in minute amounts present in a single sample. It can also identify viral RNA by including a pre-processing step that converts RNA to DNA using reverse transcriptase [2, 6, 22].

2.4.2 Reverse transcriptase-PCR (RT-PCR)

It was designed with the intent of amplifying RNA targets. This technology converts viral ribonucleic acid (RNA) to complementary DNA (cDNA) and then amplifies it using a standard polymerase chain reaction (PCR). It has a specificity of 99.9 to 100% and a sensitivity of 73 to 100% for detection of viral infections [2].

2.4.3 Quantitative real-time PCR (qPCR)

Amplification and detection of nucleic acids of the virus are carried out concurrently in this technique. The amplification product is detected based on the quantity of light emitted by the specimen. A thermal cycler is used to measure the specimen’s fluorescence emission. The computer, coupled with thermal cycler via appropriate software, records the information and generates an amplification graphic for each reaction cycle [2, 37].

2.4.4 Real-time quantitative reverse transcription PCR (RT-qPCR)

RT-qPCR, short form for Real-Time Quantitative Reverse Transcription PCR, is a powerful molecular technique that allows for accurate detection and measurement of the amplified product during each cycle of the PCR process. This method became possible with the development of an oligonucleotide probe designed to hybridize specifically with the target sequence. The key to RT-qPCR lies in the use of a target-specific probe, which, during the PCR process, undergoes cleavage due to the 5′ nuclease activity of Taq polymerase. This cleavage occurs only when the probe binds specifically to the targeted sequence, enabling precise and real-time monitoring of the amplification progress. To initiate this process, reverse transcriptase is employed to convert messenger RNA (mRNA) or total RNA into complementary DNA (cDNA). This conversion step is crucial because it allows RNA, which is the intermediate in gene expression, to be converted into DNA, which is more stable and amenable to PCR amplification. Once cDNA is obtained, the qPCR process is carried out using it as the template. During qPCR, the number of copies of the target DNA sequence is exponentially amplified with each cycle, and the fluorescence emitted by the cleaved probe is continuously measured. This fluorescence data is then used to determine the quantity of targeted DNA present in the initial sample, allowing for accurate quantification (Figure 16) [37].

Figure 16.

Diagrammatic representation of two-step RT-qPCR.

2.4.5 Restriction fragment length polymorphisms (RFLPs)

DNA viruses do not have segmented genomes. However, using the appropriate restriction enzymes, it is possible to segment the isolated viral DNA into specific segments for electrophoresis-based RFLP analysis of the various viral strains and variations. Examples: Influenza A virus can be diagnosed by this method (Figure 17) [38].

Figure 17.

Diagrammatic representation of restriction fragment length polymorphism analysis of viral DNA.

2.4.6 Nucleic acid sequence-based amplification assay (NASBA)

The nucleic acid sequence-based amplification (NASBA) process consists of two stages: denaturation followed by temperature labile polymerase-dependent isothermal amplification. Fluorochrome-labeled probes are introduced for real-time observation. Researchers developed multiplex RT-NASBA (Real-Time Nucleic Acid Sequence-Based Amplification) to simultaneously detect Norovirus genogroup II, Astrovirus, and Rotavirus A. Compared to RT-PCR, RT-NASBA offers several advantages. Since it is performed under isothermal conditions, there is no need for cooling and heating, and the use of reverse transcriptase as a separate step is not required. As a result, amplification is considerably faster, and both the sensitivity and specificity of RT-NASBA are higher than RT-PCR. In the working principle of RT-NASBA, the process begins with reverse transcriptase using antisense primer P1, carrying a 5′-overhang T7 polymerase promoter sequence, to produce cDNA. RNase H then hydrolyzes the RNA in the RNA: cDNA hybrid, resulting in single-strand cDNA. In the next step, with the help of reverse transcriptase, the sense primer P2 anneals to the cDNA, generating double-stranded DNA (dsDNA). Binding of T7 RNA polymerase to the promoter site of the dsDNA ends the non-cyclic phase and produces copies of antisense-RNA of the target RNA. During the cyclic phase, primer P2 binds first to the antisense RNA molecule, forming an RNA-cDNA hybrid through reverse transcriptase. Subsequently, RNase H hydrolyzes the RNA, and primer P1 binds to it, producing dsDNA. The process continues as T7 RNA polymerase generates new RNA copies, which serve as templates for further amplification. Throughout the amplification, a molecular beacon continuously keeps in check about the RNA product accumulation in real time. The advantages of RT-NASBA are direct amplification of RNA, high amplification efficiency with exceptional specificity, real-time detection capabilities, and reduced risk of contamination. This technique can be used to detect viruses such as Rotavirus A, Norovirus genogroup II, and Astrovirus (Figure 18) [39].

Figure 18.

Diagrammatic representation of the working of nucleic acid sequence-based amplification technique.

2.4.7 DNA microarrays

This method is having a great impact in the field of detection of nucleic acid of viruses. In DNA microarray diagnosis, a test sample containing fluorescently tagged viral nucleic acids is utilized. The microchip is composed of a solid support matrix with “printed” areas containing several hundred to several thousand distinct oligonucleotides. They are virus-specific. Fluorescence-based detection is used to detect and quantify the outcomes of hybridization between immobilized probes and fluorescently tagged target sequences [2]. Examples: Parainfluenza 3, Poliovirus 1, Respiratory syncytial virus can be diagnosed by this method (Figure 19) [40].

Figure 19.

Diagrammatic representation of DNA microarray technique.

2.4.8 Next-generation sequencing (NGS)

It is a revolutionary approach that allows for direct sequencing of DNA fragments without the requirement for cloning. Bioinformatic tools are used to analyze the sequencing data. For instance, pyrosequencing Roche 454 identifies the pyrophosphate release after nucleotide is incorporated into a DNA polymerization process. Illumina’s next-generation sequencing technologies detect the release of fluorescence from nucleotides inserted during the DNA polymerization process. By measuring the ionic current of RNA/DNA molecules flowing through the nanopores, the Oxford nanopore (MinIon) technology sequences the target nucleic acid [2, 22]. Examples: Porcine circovirus 3, Sparrow Deltacoronavirus, Porcine pegivirus, Porcine nordavirus, Influenza D virus can be diagnosed by this method (Figure 20).

Figure 20.

Diagrammatic representation of next-generation sequencing technology. (A) Emulsion PCR. (B) Solid-phase amplification. (C) Sequencing and imaging. (D) Data analysis using the software that is available or an integrated workflow such as the GATK pipeline.

2.4.9 CRISPR-Cas system-based determination of viral infections

Rapid nucleic acid detection plays a crucial role in various human health and biotechnological applications, including identification of infectious diseases, pathogens in agriculture, and disease-associated circulating DNA or RNA. In this context, CRISPR-Cas-based methods are gaining attention for diagnosing infectious and non-infectious disorders. These innovative approaches have the potential to outperform traditional PCR-based techniques in numerous applications. The CRISPR-Cas system relies on Cas effector proteins that can cleave DNA or RNA. Guide RNAs direct Cas proteins to the specific target site for nucleic acid cleavage. By modifying the guide RNA, researchers can easily target specific sequences, making it a valuable tool for nucleic acid detection. Three primary CRISPR-Cas methods have been proposed: SHERLOCK, DETECTR, and HOLMES [41].

In the type II CRISPR/Cas system, three phases include CRISPR acquisition, crRNA biogenesis, and interference with invasive DNA. Cas nucleases cleave the RNA or DNA at the identification site and some Cas proteins can also cause collateral cleavage (off-target cutting). This collateral cleavage phenomenon can be utilized for nucleic acid detection. Targeted nucleic acids are initially amplified using an isothermal process, often RPA, to aid Cas-mediated detection. In the CRISPR-Cas system, guide RNA specific to the target DNA location is introduced. When guide RNA binds with target DNA, it leads to target cleavage along with collateral cleavage. The collateral activity of the DNA-recognizing Cas protein, Cas12a, cleaves a fluorophore probe, generating a detectable signal in the test. Detection can be achieved through fluorescence measurement or by using a lateral flow strip for quicker results. Among various CRISPR-Cas techniques, the FELUDA test stands out as a breakthrough technique developed in India [42]. This test utilizes the revolutionary CRISPR technology to detect the SARS-CoV-2 virus. The FELUDA test involves RNA extraction and one-step RT-PCR to convert viral RNA to DNA and amplify it. A mixture containing Cas9 protein, guide RNA, and amplified viral DNA is then created. The Cas9 protein, guided by the RNA, specifically binds to the matching viral DNA, forming a complex. A piece of paper, similar to a home pregnancy test, is used for lateral flow detection, showing a single line for a negative result and a double line for a positive result within 45 minutes. Although the FELUDA test offers rapid results, the initial RNA extraction and RT-PCR steps must be performed in a laboratory, preventing it from being conducted at home (Figure 21) [43].

Figure 21.

Diagrammatic representation of CRISPR Cas technique. Nucleic acid detection in (A) SHERLOCKv2, (B) SHERLOCK, (C) DETECTR, and (D) FELUDA assays using CRISPR-Cas technology.

2.4.10 Mass spectrometry

Mass spectrometry has been successful at significantly improving the diagnosis of many diseases. Present-day mass spectral techniques were first developed by Aston and Dempster in the 1900s. For viral diagnosis at the cellular level, they were applied. A combined assay of multiplex PCR and MALDI-TOF MS was developed to detect multiple viruses simultaneously in a single sample. PCR combined with mass spectrometry is better as compared to standard methods in cases where multiple viruses are present [44]. Example: SARS-COV-2 can be diagnosed by this method (Figure 22) [45].

Figure 22.

Diagrammatic representation of mass spectrometry.

2.4.11 Biosensors

Biosensors allow rapid virus detection in a wide range of samples. The selection of the appropriate membrane forms an important part of biosensor construction. Many studies have employed organic and inorganic materials such as NaN3, nitrocellulose, nylon, and polyether-sulfonate. The detection limit is the primary goal of all biosensors, regardless of whether the technique of detection is the full virus or any protein. Molecular motor biosensor based on F0F1-ATPase, a molecular motor created by using a biotin-streptavidin method to attach the probe to F0F1-ATPase, is one of the biosensors for the detection of viruses. Detection of food-borne enteric viruses can be done in less than an hour by this technology [46]. For the identification of enteric pathogens, a photoluminescence-based immuno-biosensor was built with a graphene oxide array and the detection of Rotavirus was done using the fluorescence resonance energy transfer (FRET) method [47].

It has high sensitivity and relies on specific complementary single-stranded DNA target sequences being hybridized by DNA probes mounted on gold nanoparticles. The main benefit of this method is that its efficiency is unaffected by low ion and molecule concentrations. Its reference database must be updated on a regular basis, and it can be insufficiently sensitive for detection of every mutation. Gold electrode covered by high-affinity binding peptide of Norovirus, which is highly sensitive, was developed by Baek et al. [48]. Enteric viruses can be detected on-site by using an electrochemical biosensor because they are easier to detect and have a high sensitivity. In case of endemic breakout, it is ideal for mass deployment due to its easy usage. Examples: Respiratory Syncytial virus, Influenza virus, and SARS-COV-2 can be diagnosed by this method (Figure 23) [49].

Figure 23.

Diagrammatic representation of working of biosensors.

2.4.12 Aptamers

Aptamers are a stretch of peptides or oligonucleotides that have got a lot of consideration from researchers and scientists all around the world as an alternative to antibodies. They have all of the characteristics of antibodies and are also thermostable and cost-effective, making them ideal for usage in resource-constrained contexts [50]. Aptamers are combined with RT-PCR for successful diagnosis, and utilized as Aptamer-linked immunosorbent assays, cantilever-based aptasensors, and SPR-based similar to ELISA. Due to their very high sensitivity and portability, ease of use, and rapid detection, aptasensors offer the potential for use in point-of-care detection systems [51]. In situ, capture RT-qPCR technique-based aptamers have recently been developed for the identification of human noroviruses from clinical samples [52]. Examples: Human norovirus, Dengue virus, Ebola virus, etc., can be diagnosed by this method (Figure 24) [53].

Figure 24.

Diagrammatic representation of working of Aptamers. (A). Aptamer library is created and attached to target protein. (B). Protein with its specific aptamer is selected and non-specific aptamer is removed by washing. (C). Aptamers bounded with protein are eluted and purified. (D). Characterization of selected aptamer is done. (E). This unbound Aptamer is then amplified for creation of library. (F). Unbound Aptamers are single-stranded DNA, and it is converted into double-stranded DNA after final selection. (G). After conversion, next-generation sequencing is done. (H) Negative selection is done using negative control protein; if unbound aptamers bind to negative protein, then it is discarded.

2.4.13 Surface Plasmon resonance (SPR) spectroscopy

Surface plasmon resonance (SPR) spectroscopy is a good method to monitor non-covalent chemical interactions in real time and without any interference. It serves as a label-free assay, producing a visible or fluorescent signal without the need for tags, dyes, or specialized reagents like enzyme-substrate complexes. This attribute makes SPR an excellent tool for studying diverse non-covalent interactions, such as those between proteins and DNA, proteins and cells, RNA and DNA, proteins and carbohydrates, receptor-inhibitor complexes, proteins and peptides, and self-assembled monolayers. SPR has found extensive applications in various fields, including clinical immunogenicity studies, drug discovery, and ligand fishing. The principle of SPR involves a light beam passing through a prism and interacting with a metal surface, typically gold or silver. At the interface between these two media, plasmons, or electron charge density waves, are generated, facilitating the real-time monitoring of interactions occurring at the surface. This unique capability of SPR makes it a valuable technique for investigating molecular interactions in diverse research areas [54].

When the metal surface (usually gold or silver) is hit by polarized light at the interface of two mediums with different refractive indices, Surface plasmons, also known as oscillations of free electrons on the metal surface, are excited and detected by SPR techniques. With the Kretschmann-Raether setup, light is focused via a prism onto a metal surface, where the reflection of light is measured. The metal surface absorbs light as the plasmons are set to resonate at a specific incidence angle or resonant angle. This causes a decrease in reflection strength and a dark band to appear in the detector. Most often, proteins are immobilized on the metal surface, using a series of flow cells, and prospective ligands are then introduced over the surface. The resonant angle of the least intensity of light reflected during these biomolecular interactions is identified. The molecules’ binding causes this resonant angle to alter. The SPR sensorgram records these interactions in real time. Its utility has surpassed that of conventional viral detection techniques, particularly in healthcare and medical diagnosis. With the identification of antibodies against various antigens found in the virus, SPR studies were used to identify different Epstein-Barr virus phases. SPR was also used to monitor binding kinetics of dengue antibody with corresponding dengue antigen in real time. Yet, major barriers to reaching the full potential of SPR include the need for sophisticated infrastructure and trained personnel (Figure 25) [55].

Figure 25.

Diagrammatic representation of surface plasmon resonance.

2.5 Other nucleic acid isothermal amplification methods

Other PCR variants do not need high temperatures (95°C) for denaturing the DNA and binding of new primers. These techniques depend on enzymes to displace the strand, including Reverse Transcriptase-PCR (RT-PCR), Nucleic Acid Sequence-Based Amplification (NASBA), Polymerase Chain Reaction (PCR), and Quantitative Real-Time Polymerase Chain Reaction (qPCR) among others. These techniques may have the advantage of being incredibly rapid and requiring no thermal cycling equipment [2].

2.5.1 Loop-mediated isothermal amplification assay (LAMP)

The loop-mediated isothermal amplification assay (LAMP) method efficiently detects DNA and RNA viruses in human specimens with high sensitivity, specificity, speed, and cost-effectiveness. This approach involves the use of at least six distinct primers (B3, F3, FIP, BIP, LB, LF) and a DNA polymerase with strand-displacement activity. By incorporating reverse transcriptase, RNA targets can also be amplified [2]. LAMP’s targeted amplification relies on multiple primers recognizing 6–8 unique sections of the target DNA, creating a loop structure that facilitates successive rounds of amplification. Viruses detected by LAMP technique include Foot and mouth disease, Norovirus, Human Papillomavirus, Cytomegalovirus, and Human immunodeficiency virus (Figure 26) [56].

Figure 26.

Diagrammatic representation of loop-mediated isothermal amplification assay (LAMP).

2.5.2 TaqMan probe-based insulated isothermal PCR

In this Rayleigh-Bénard convective PCR method, annealing, denaturation, and extension occur in separate zones in the cylindrical tube at a fixed temperature at the bottom. Vessels are insulated using jacket insulators to reduce heat loss. At the bottom of the vessel, DNA denaturation occurs; in the upper zone, annealing of primer occurs; and in the center, extension occurs. This method is simple, inexpensive, rapid, and sensitive to be utilized as a reliable diagnostic tool. Fluorescently tagged probes are included with the primers to allow for real-time monitoring of the amplification. This technique has been used to detect the CPV-2 virus (Canine Parvovirus-2) (Figure 27) [57].

Figure 27.

Diagrammatic representation of Taqman probe-based amplification technique.

2.5.3 Recombinase polymerase amplification assay (RPA)

Binding proteins and bacterial recombinase enzymes are used in recombinase polymerase amplification, which is usually done at a slightly higher temperature than normal to avoid non-specific amplification. It has revolutionized nucleic acid detection and is a good alternative to PCR. This technique requires only half an hour and no requirement for the template to be thermally denatured. Thermal annealing and denaturation are not used in RPA. Three essential proteins, single-stranded nucleic acid binding recombinase, strand-displacement DNA polymerase, and a single-stranded DNA binding protein are needed for RPA processes [58, 59]. The recombinase binds to a primer that is roughly 30–35 nucleotides long to begin the amplification reaction, after which, in the double-stranded DNA template, the complex searches for the target site. After the identification of the site by complex, it starts a chain exchange reaction right away to create a D-shaped loop. The shifted DNA chain is then bound by a single-stranded binding protein to stop primer dissociation. Recombinase-primer complex causes active hydrolysis of ATP, leading to change in the complex’s structure. The strand-displacement DNA polymerase identifies the 3′ end of primer after separation of recombinase from the nucleoprotein filament. DNA amplification reaction occurs when DNA polymerase inserts the matching nucleotide into the primer’s 3′ end, following the template sequence. With both forward and reverse primers, the amplification response can happen simultaneously in both directions. A fresh template can be created using the synthesized amplicon to accomplish exponential amplification [60]. But still, there are some shortcomings; to prevent smearing caused by the inclusion of additional components, RPA products typically need to be purified before agarose gel electrophoresis. The design and screening of RPA primers and probes cannot be done using specialized software. Therefore, time-consuming and expensive synthesis and screening experiments are needed. Furthermore, the RPA reaction is incompatible with traditional real-time PCR probes (such TaqMan probes), as they are susceptible to give false-positive results. Due to its isothermal amplification characteristics and use of time threshold rather than a cycling threshold, managing real-time amplification with RPA is difficult. As a result, original circumstances, incubation temperature, and mixing procedures all affect the responses [61]. Apart from that due to its high fidelity, portability, affordability, simplicity, sensitivity to inhibitors, and ease of use, this technique is good [62]. Examples: Influenza A virus, Dengue virus, West Nile virus, etc., can be diagnosed by this method (Figure 28).

Figure 28.

Diagrammatic representation of working of recombinase polymerase recombination technique.

2.5.4 Single primer isothermal amplification (SPIA)

In this technique, RNA functions as a primer in a chimeric DNA/RNA sequence, when RNase H degrades the 3′ end of RNA. And to stop the reaction, blockers are used. After sequence amplification, a short-oligonucleotide sequence is used as a blocker to stop further extension. RNase H digests the RNA sequence during amplification and the primers take the vacated position in the following round. SYBR Green II is employed for the identification process when the amplified product binds to single-stranded DNA. Even 1 ng of mRNA can be amplified using this approach. The major drawback is that because RNase H prevents the reaction from being amplified, and it must be confirmed on agarose gel electrophoresis. This method is excellent when it is not possible to design a robust primer [63]. RNase H, a DNA polymerase with high strand-displacement activity, and a single target-specific chimeric primer of ribonucleotides at its 5′ end and deoxyribonucleotides at its 3′ end are all used in SPIA. In the target DNA molecule, chimeric primer hybridizes to a complementary sequence for the amplification process to begin. The target DNA strand is extended by DNA polymerase, beginning with the hybridized primer. When the primer extension begins, RNase H cleaves the 5′RNA part of extension primer (DNA-RNA hybrid), so that the primer binding site left in the target DNA strand can bind to the new chimeric primer. The newest binding primers compete with previously extended primers to control binding to the complementary target DNA sequence. DNA polymerase binding stabilizes the newly attached primer and removes the 5′ end of the previously extended one. When the primer extension restarts replication, 5′ of RNA component of the newly extended primer is cleaved by RNase H, so that the primer binding site is freed for the next replication cycle. Up to 10,000 replications are initiated and repeated using the original transcripts. With this method, amplification of many nucleic acid types is possible [64, 65]. Examples: Bovine Coronavirus can be diagnosed by this method (Figure 29) [66].

Figure 29.

Diagrammatic representation of working of single primer isothermal amplification.

2.5.5 Polymerase spiral reaction (PSR)

The PSR approach was invented lately and was first applied to E.coli BL-21 cells for amplification of blaNIM–1 plasmid. It has rapidity, high specificity, and efficiency under isothermal condition with requirement of one pair of primers and an enzyme. The resemblance of this technique is like a traditional isothermal PCR that is done at a fixed temperature of 61–65°C with a few tweaks to the primer design. The 5′ end of primers has an exogenous area that is 22–25 bases long in both forward and reverse directions. One primer’s exogenous region is oriented in the opposite way as the other’s exogenous region. Using turbidity and/or fluorescent dye (SYBR Green)-based imaging, the amplification of this reaction can be observed in real time and results in the formation of a complex spiral structure. This technique is a cost-effective and convenient alternative for clinical screening, primary quarantine purposes, and on-site diagnosis [67]. Examples: West Nile virus [68] and Porcine circovirus type 3 [69] can be diagnosed by this method (Figure 30).

Figure 30.

Diagrammatic representation of working of polymerase spiral reaction technique.

2.5.6 Quantitative polymerase chain reaction (qPCR) based on primer-probe energy transfer

It utilizes Förster resonance energy transfer (FRET) for simultaneous detection and quantification of viral genome copies. Primer-probe energy transfer (PriProET) concept is employed, enabling the detection of multiple viral variants simultaneously. PriProET offers flexibility in detecting various target nucleic acids, even with changes in the targeted regions and mutations in the viral genome [70]. In this method, the probe sequence is 25–30 nucleotides long, and quencher dye (TAMRA) is used to tag 3′ end, and fluorescent reporter dye (FAM) is used to tag 5′ end [71]. TaqMan probes are designed to anneal within a region of DNA that has been amplified by a particular set of primers. To improve their binding affinity to the target sequence, minor groove binder (MGB) moiety was conjugated with TaqMan probes. The Taq polymerase’s 5′ to 3′ exonuclease activity breaks down the probe, which has annealed with the template as it extends the primer and creates the nascent strand. The fluorophore is released from the probe during degradation, which also breaks the probe’s proximity to the quencher, and quenching effect is eased and fluorescent from fluorophore is released. As a result, DNA template amount present in the PCR and the amount of fluorophore emitted precisely correlate with the fluorescence detected in the quantitative PCR thermal cycler. Examples: Respiratory syndrome virus, Hepatitis E virus, Feline coronavirus [71], snd Porcine coronavirus [72] can be diagnosed by this method (Figure 31).

Figure 31.

Diagrammatic representation of working of real-time polymerase chain reaction (qPCR) based on primer-probe energy transfer.

2.5.7 Hybrid capture assay

In this nucleic acid hybridization assay, the samples containing the target DNA are hybridized with Human Papillomavirus RNA (HPV RNA) probe. The formed RNA-DNA hybrids are captured using a microplate coated with DNA-RNA hybrid antibodies. Detection is achieved through chemiluminescence. To detect the hybrids, specific antibodies for DNA-RNA hybrids are coupled with alkaline phosphate, and they react with the hybrids on the microplate. This leads to the generation of a chemiluminescent signal when a chemiluminescent substrate is applied. The signal is amplified as multiple conjugated antibodies bind to each hybrid. The chemiluminescent substrate is then cleaved by alkaline phosphate, resulting in the emission of light. This emitted light is measured with a luminometer, which enables the presence of target DNA to be determined. Example: Human Papillomavirus can be diagnosed by this method (Figure 32) [73].

Figure 32.

Diagrammatic representation of hybrid capture assay.

2.5.8 Proximity ligation assay (PLA)

Proximity ligation assay is a sensitive, homogeneous, and highly specific immunohistochemical technique that combines the sensitivity of PCR with the specificity of ELISA. It has been demonstrated that this method, which was created by Fredrickson and colleagues in 2002, overcomes the challenges associated with trying to visualize and study individual proteins, protein-protein interaction, and post-translational modifications, such as glycosylation, acetylation, and phosphorylation. PLA technology enables the identification of endogenous protein interactions. The detection of protein proximity is the basis for this approach. PLA employs a single set of primary antibodies. These primary antibodies are directed toward proteins of interest, such as two separate proteins or the two epitopes of fusion proteins, whose proximity we want to investigate. Secondary antibodies attached to short DNA oligonucleotides are used to identify primary antibodies generated in various species. The DNA strands hybridize and participate in the rolling circle synthesis of DNA, if the oligonucleotides are close together (theoretically less than 40 nm). Hybridization of fluorescent-labeled oligonucleotides can also be used to detect these DNA copies. The resulting high fluorescence concentration may be easily viewed and quantified under a microscope. Each dot represents one colocalization [74]. Proximity Ligation Assay can be done using either a direct or indirect method. The Ab-Oligo conjugates are used by direct method and the indirect method uses primary antibodies that have not been modified and are identified by secondary Ab-Oligo conjugates. The method can make use of either polyclonal, monoclonal, or a combination of both antibodies. Example: SARS COV-2 can be diagnosed by this method (Figure 33) [75].

Figure 33.

Diagrammatic representation of proximity ligation assay. (A). Direct proximity ligation assay uses antibody pair with primary conjugation. (B). Indirect proximity ligation assay uses secondary antibody conjugate.

2.5.9 Helicase-dependent amplification (HDA)

The Helicase-dependent amplification is the simplest method for isothermal nucleic acid amplification, it uses a helicase to unwind DNA duplexes isothermally rather than heat, in PCR, mimicking the in vivo process of DNA replication. The original HDA system needs two important proteins, single-stranded DNA binding protein (SSB) and MutL, to maintain ssDNA and prevent the complementary strands from binding again. In a method very similar to the PCR without thermal cycling, at 3′ end and 5′ end of the targeted DNA, two oligonucleotide primers hybridize after DNA has been isothermally unwound and stabilized. When deoxynucleotide triphosphates are added, DNA polymerase extends annealed primers and double-stranded amplified product is produced. The target dsDNA is amplified exponentially as the procedure is repeated. HDA needs only two primers and a few proteins to perform; it has a simpler assay method than other isothermal reactions like LAMP. HDA may be utilized with unprocessed samples without the necessity for the extraction of DNA or RNA. Lateral flow-based detection and fluorescent dyes can be used for the detection of amplified products. Examples: Herpes simplex virus, Human immunodeficiency virus, and Human Papillomavirus can be diagnosed by this method (Figure 34) [76].

Figure 34.

Diagrammatic representation of helicase-dependent amplification.

2.5.10 Rolling circle amplification (RCA)

The replication of circular DNA occurs naturally and rolling circle amplification (RCA) imitates this process. In contrast to procedures like helicase-dependent amplification and recombinase polymerase amplification, which amplify linear DNA, this procedure is designed for use in the amplification of circular DNA sequences. In its most basic form, RCA just needs one primer, which is followed by the strand-displacing polymerase that starts replication. The circular DNA repeat sequences make up the linear product known as the amplicon, which is the result. Although exponential amplification is not possible with this RCA technique, the procedure can be changed to allow for rapid, exponential amplification. These methods include using the two-primer hyper-branched RCA technique and another alternative method called multiple-primed RCA, which uses several primers and padlock probe-based RCA, and when desired target is single-stranded linear DNA, these types of probes are used. Either fluorescence or intercalating dyes can be used to detect the end products in real time. The assay design for RCA is relatively simplistic, requiring at most one primer to amplify circular DNA or RNA and not denaturing the target. It can be used for the sequencing of emerging strains, rapid viral genome amplification, and variation analyses between two variants in addition to viral diagnostics [54]. Examples: Ebola virus, Zika virus, Dengue virus [77], SARS-COV-2, Influenza A and B, Papillomavirus type 16, and Human immunodeficiency virus can be diagnosed by this method (Figure 35) [78].

Figure 35.

Diagrammatic representation of rolling circle amplification.

2.5.11 Signal-mediated amplification of RNA technology (SMART)

The three-way junction formation is the foundation of this technology (3WJ). No thermal cycler or target sequence copying is needed; this technique depends on Signal amplification. It produces a signal that is suitable and extremely target-dependent to find targets in DNA or RNA. The SMART has two components, for extension matching single-stranded oligonucleotide probe with a zone defined on target for hybridization of each probe and the other considerably hybridization of shorter region at adjacent spots to the opposite probe. Annealing of two probes with each other in the presence of a specific target forms 3WJ structure. Extension of short probe by Bst DNA polymerase after 3WJ formation by replication of opposite template probe produces double-stranded T7 RNA polymerase promoter sequence. To enable the synthesis of 3WJ, the generated promoter enables T7 RNA polymerase for synthesizing multiple copies of RNA amplicon in the presence of a particular target. Each RNA amplicon has the potential to self-amplify when it binds to second template oligonucleotide (probe for the amplification), and a double-stranded promoter is created when extended by DNA polymerase. Due to this, there is an increase in transcription; as a result, RNA amplicons increase, which can be detected in a real-time format or by Enzyme-linked Oligosorbent Assay (ELOSA). The target sequence is not itself amplified throughout this process, which is a signal amplification technique (Figure 36) [65].

Figure 36.

Diagrammatic representation of signal-mediated amplification of RNA technology (SMART).

There are many diagnostic methods for the diagnosis and detection of human viruses. These diagnostic methods have changed drastically in past decades, but still, there is a gap which leads to wrong interpretation of results. The main reasons for these misinterpretations are cross reactivity, loss of genomic material, contamination of samples, multiple viruses causing same disease, etc. and symptom shown at later stage. However, the advanced techniques have greatly improved the diagnosis of these human viruses but they are cumbersome with lengthy procedures and need expertise. But before detection, awareness and knowledge regarding diseases caused by human viruses are most important. It will become easier to diagnosis these viruses. With proper knowledge and advanced techniques, we can come up with more new handy techniques for the detection of human viruses (Figure 37).

Figure 37.

Image credit: “Prevention and treatment of viral infections”, by OpenStax college, biology, CC BY 4.0. Modification of original work by Mikael Häggströms.

Advertisement

3. Advantages of recent methods

High sensitivity and specificity; combination of different assays in one method; automation of assay; requirement of small sample assay; less time-consuming; rapid and low detection limit; multiplex reaction; rare drug detection; use of next-generation sequencing; low reaction cost; lower risk of contamination.

Advertisement

4. The future for viral diagnosis

All detection technology will advance toward more sensitive and less exhaustive methods of disease detection. A greater emphasis should be placed on molecular and immunological diagnosis to facilitate and expedite diagnosis. Viruses evolve at an alarming rate, making them extremely difficult to diagnose and identify. In the near future, a quicker diagnostic tool may be developed. Clinical virology is undergoing a paradigm shift as a result of new diagnostic tools. Molecular diagnostic techniques are more sensitive and specific. However, technological capabilities alone are insufficient; health promotion programs should emphasize the need for early detection of infectious disease outbreaks and their dissemination. Less developed countries are unable to advertise and cannot afford pricey diagnoses; this issue must be resolved by providing all assistance necessary to halt the spread of disease. It is envisaged that in the near future, new high-quality tests will be affordable enough for low-income countries to afford and implement policies for disease control and prevention.

Advertisement

5. Conclusion

For virus detection, a combination of molecular and serological methods should be employed. Having a thorough understanding of the disease’s pathophysiology and epidemiology will only aid in determining the best course of action for treatment. Emerging and re-emerging viruses should be identified using new approaches. Viral DNA or RNA is detected using molecular techniques far before an antibody reaction is triggered. Viral infection diagnosis and treatment rely heavily on immunoassay; however, these tests are not generally available. Since only wealthy nations can currently afford good-quality diagnostics, the application of cutting-edge techniques in poor nations and disease-endemic regions is now being held up. It is projected that attempts will continue to be made to provide new, low-cost, high-quality tests, which will significantly strengthen disease control tactics. The majority of emerging nations have built necessary departments and institutes to train their nationals both domestically and overseas. There is hope that efforts will continue to be made to provide new inexpensive high-quality test for low-income nations, which would drastically boost their populations’ disease control tactics (Tables 35).

VirusesDiseasesFamily/GroupDiagnostic methodsRefs.
RNA viruses
AichivirusAcute gastroenteritisPicornaviridaeTaqMan RT-qPCR method[79]
ArenavirusLymphocytic choriomeningitisArenaviridaeSerological method, Culture[80]
CosavirusMeningitis, EncephalitisPicornaviridaeRT-qPCR method[81]
CoxsackievirusAseptic meningitis, Hand, foot, and mouth diseasePicornaviridaeELISA, RT-PCR[82]
Dengue virusHemorrhagic fever, Shock syndromeFlaviviridaeELISA, Complement fixation, PCR[83]
Ebola virusEbola Hemorrhagic feverFlioviridaeELISA, RT-PCR, Biosensors, RT-LAMP, Immunohistochemistry,[84]
EnterovirusMuscle paralysis, Inflammation of brainPicornaviridaePattern Recognition Receptor, Culture[85]
Hepatitis virusHepatitis, JaundicePicornaviridaeHistology, Serological methods, Molecular methods[86]
Human AstrovirusGastroenteritisAstroviridaeELISA, RT-PCR, Electron microscopy, Immunodetection[87, 88]
Human immunodeficiency virus (HIV-1/2)AIDSRetroviridaeImmunoassay, RT-PCR, Western blot[89]
Human PicobirnavirusAcute gastroenteritis, diarrheaPicobirnaviridaeRT-PCR[90]
Human Rhino virusCommon cold, upper respiratory tract illness, bronchiolitisPicornaviridaeMolecular method, ELISAs, complement fixation, culture[91]
Human Rotavirus ADiarrheaReoviridaeELISA, RT-PCR, Hybridization (DIG) probe method, RT-LAMP, Raman spectroscopy, paper-LAMP, Culture[92, 93, 94]
Human SapovirusAcute gastroenteritisCaliciviridaeRT-PCR method[95]
Influenza virus(H1N1)Swine fluOrthomyxoviridaeFluorescent immunochromatographic strip test (FICT), RT-qPCR[96]
Japanese encephalitis virusAcute encephalitis syndrome, mental retardationFlaviviridaePRNT, RT-PCR, hemagglutination inhibition, virus neutralization assay, Complement fixation, ELISA[97, 98]
LyssavirusRabiesRhabdoviridaeRT-PCR, fluorescence antibody staining, serological method, culture[99]
Mammalian orthoreoviruses 3Pneumonia, encephalitis, respiratory distress syndrome, jaundice, alopeciaReoviridaeRT-PCR, electron microscopy[100, 101]
NorovirusSevere vomiting, diarrheaCaliciviridaeEIA, RT-PCR, Aptamer-based in situ capture, RT-qPCR, electrochemical biosensor[46, 50, 92, 102]
Parainfluenza virusOtitis media, pharyngitis, conjunctivitis, tracheobronchitisParamyxoviridaeColorimetric enzyme-linked immunoassays, PCR, complement fixation Culture[103]
ParamyxovirusMumpsParamyxoviridaeFocus reduction neutralization test (FRNT), ELISA test, RT-PCR, RT-LAMP[104, 105]
PoliovirusPoliomyelitisPicornaviridaePCR, virus isolation[106]
Respiratory syncytial virusBronchiolitis, pneumonia, asthma, lung hyperinflationPneumoviridaeEnzyme immunoassay, immunofluorescent assay[107]
Rubella virusMeaslesParamyxoviridaeReverse transcriptase PCR, enzyme immunoassay[108]
SalivirusAcute gastroenteritisPicornaviridaeRT-PCR[109]
Severe acute respiratory syndrome coronavirus 2 (SARS-COV-2)Coronavirus disease-19 (COVID-19)CoronaviridaeReal-time RT-PCR, ELISA test, lateral flow immunochromatographic assay (LFIA), pyrosequencing, RT-LAMP, CRISPR-Cas, nanomaterials-based SARS-CoV 2 detection.[110, 111]
TorovirusDiarrhea, pyrexia, lethargyTobanviridaeRT-LAMP and RT-PCR[112]
West Nile virusFever, encephalitis, meningitisFlaviviridaeELISA, RT-PCR[113]
DNA viruses
AdenovirusViral meningitis, gastroenteritis, conjunctivitisAdenoviridaeRT-PCR, antigen detection, ELISA, immunochromatography, culture[114, 115]
Epstein-Barr virusMononucleosis, Burkitt lymphoma, hemophagocytic lymphohistiocytosis, Hodgkin’s lymphomaHerpesviridaeFluorescent antibody staining, serological method[116]
Herpes simplex virusOral herpes, genital herpesHerpesviridaePolymerase chain reaction, fluorescent antibody staining[117]
Human Bocavirus 1Bronchiolitis, pneumoniaParvoviridaeEIA, ELISA, RT-qPCR,[118]
Human BufavirusGastroenteritis, diarrheaParvoviridaeRT-qPCR, TaqMan real-time PCR[119, 120]
Human PapillomavirusCervical, oropharyngeal cancerPapillomaviridaeELISA, qPCR, hybrid capture assay, in situ hybridization, high-throughput pseudovirion-based neutralization assay (HT-PBNA)[121, 122]
PoxvirusSmallpox, cowpox, monkeypoxPoxiviridaeHistology, ELISA, immunodiffusion assay, immunochemistry, and immunofuorescence assay, RFLP, qPCR, neutralization test, Western blot assay, Hemagglutination inhibition assay[123]
Varicella Zoster virusChickenpoxHerpesviridaeELISA, PCR, direct immunofluorescence[124]

Table 3.

Some common viruses and their diagnostic methods.

VirusPCR analysisReference
Sensitivity (%)Specificity (%)
SARS-CoV-2 (Coronavirus)9595[12]
Cytomegalovirus8298[125]
Dengue virus88.9100[126]
Epstein-Barr virus9798[125]
Hepatitis virus9198[127]
Herpes simplex virus10099.5[125]
Human Immunodeficiency virus91.6100[128]
Japanese encephalitis virus9697[129]
Lyssavirus96.692.7[130]
Rubella virus100100[131]
Varicella Zoster virus90.599[132]
West Nile virus94100[133]

Table 4.

Sensitivity and specificity.

Diagnostic methodsPrincipleAdvantageDisadvantageExamples
Virus isolation (VI)Virus samples are grown on cell lines and tested for their ability to infect, and if the cells exhibit cytopathic effect, the virus is isolated and cultured.Produces further material for study, highly sensitive.Time-consuming, expensive, not applicable for non-viable virus.Adenovirus, Cytomegalovirus, Enterovirus, Influenza, Japanese encephalitis, COVID-19.
Serological methodThese methods are used to study antigen-antibody interaction and identify them in samples.Rapid and sensitive, serotype information can be obtained, high sensitivity, readily available diagnostic kit.For almost all viruses, antigen/antibody cannot be detected, interpretation can be false, low sensitive as compared to PCR.Rubella virus, Measles, Hepatitis A, Cytomegalo virus.
Polymerase chain reaction (PCR)Amplification of small segment of genes to produce billions of copies in the in vitro condition in a short period of time.Rapid, highly sensitive, applicable to all viruses including non-cultivable, multiplexing.Contamination of DNA, detection of non-relevant co-infections due to high sensitivity, low specificity.HIV, Hepatitis B and C, Human Papillomavirus, Coronavirus.
Latex particle
Agglutination (LPA)
Formation of visible clump when antibody or antigen binds with antigen or antibody immobilized on latex particles.Less time-consuming, analytic sensitivity is high, provides consistency, uniformity, and stability, easy to interpret.False negative reaction, amount of binding influenced by pH, osmolarity, ionic concentration.Rabies, Avian influenza virus.
Radioimmunoassay (RIA)Competitive binding reaction is the fundamental tenet of RIA, where the analyte competes with an antigen that has been radiolabeled for binding to a fixed antibody or a receptor’s binding sites.High specificity and sensitivity, can detect small amount of antigens or antibodies.Radiation hazardous, high cost of waste disposal requires a specially trained person, requirement of special license.Mumps, Measles, Influenza A, Hepatitis C.
Western blotting (WB)Interaction of protein and probe or antigen/antibody for detection of protein.Can detect very small quantity of sample, uses antigens and antisera as diagnostic tool, high specificity and sensitivity.Non-intended protein can react, high amount of sample can give wrong result, appearance of bubbles while transferring.Coronavirus, HIV, Flaviviruses.
Virus neutralization test (VNT)Certain antitoxins or antibodies can lessen or neutralize a variety of biological effects brought on by various viruses or enzymes and toxins released by them.Specificity is high and measures neutralizing antibodies.Time-consuming, safety is low, needs to be operated in BSL 3/4.Rabies, Coronavirus, HIV.
Hemagglutination
Inhibition (HI)
Ability of viruses to cause red blood cells to agglutinate and agglutination is stopped by antibodies.High specificity, easy and simple to perform, no instrument required for reading result.Sensitivity is low, hemagglutinins are not present in all viruses, Interpretation often subjective.Influenza, Measles, Mumps, Rubella.
Immunofluorescence (IF)Identification of particular target antigens by using fluorescently-labeled antibodies and enables the use of fluorescent dyes to visualize the distribution of the target molecule using a fluorescence microscope.High specificity and sensitivity, easy to operate, multiple targets can be detected.Specialized trainee, cross-reactivity, long storage of immunofluorescence sample cannot be done.Influenza A and B, Coronavirus
Immunodiffusion (ID)Insoluble visible lines of precipitate in a gel are made by interaction of soluble antigens and antibody.Cheap, reliable, repeatable, indicate identity, cross reaction, and non-identity between different antigens.They are relatively insensitive and are best for detecting viral antigens or antibodies in persistent viral illnesses where viral antigens are present all of the time.Bovine leukemia virus, Avian influenza virus, Herpes virus.
Complement fixation test (CFT)Identification and measurement of antibodies in serum that do not immediately produce a precipitate or agglutinate when reacting with antigen.Interpretation of result easy, small quantity of antigen or antibody can be detected, good sensitivity.Time-consuming, labor-intensive, cross-reactivity shown, not specific.Herpes simplex virus, Rubella virus, Polyoma virus.
Nucleic acid sequence-based amplification (NASBA)It is a real-time enzymatic amplification method that can identify and amplify RNA even when DNA is present.Reaction occurs isothermally at 41°C, faster amplification kinetics, three enzyme actions can produce more than 109 copies in just 90 minutes, no risk of DNA contaminationThermolabile enzyme must be used for isothermal reaction, extraneous nucleic acids may contaminate it.H5 Avian influenza virus, Respiratory Syncytial virus, Dengue virus.
Loop-mediated isothermal amplification (LAMP)Use of DNA polymerase with strand-displacement activity is the key principle due to which dsDNA does not need to be denatured to allow for primer annealing and subsequent amplicon elongation.Rapid, sensitive, increase in specificity by using multiple primers, can be quantitative or qualitative.Requirement of increased operating temperature mainly of 65 °C; requirement of specific software for designing the primer; risk of contamination.Coronavirus, Zika virus, Dengue virus, Lassa virus, Hepatitis B.
Proximity Ligation Assay (PLA)Primary antibodies from different species are directed against two proteins of interest, then the secondary antibodies coupled to DNA probes are added, if the two proteins are close to one another, the hybridized DNA will be used for rolling circle amplification.Fast, ultra-sensitive, highly efficient, no sample purification needed, suitable for a wide range of sample types, specificity of ELISA, and sensitivity of PCR utilized to produce result.Dependent on quality of antibody used, non-specific ligation of oligonucleotide can produce background signal, time-consuming, covalent conjugation of oligonucleotide to antibody can be difficult.Foot and mouth disease virus, Influenza (H1N1).
Recombinase polymerase reaction (RPA)It is an isothermal amplification alternative to PCR. The RPA process allows for the simultaneous detection of RNA and DNA without the need for a cDNA synthesis step by the addition of enzyme reverse transcriptase.Rapid, this technique can be performed at body or room temperature, designing of primer is simple; DNA denaturation is not required before amplification.Lower processing temperature reduces the specificity.Respiratory syncytial virus, Zika virus, Influenza virus.
Rolling circle amplification (RCA)It is an isothermal enzymatic process, where a short DNA or RNA primer is amplified to yield a long single-stranded DNA or RNA using a circular DNA template and particular DNA or RNA polymerases.Availability of different formats (e.g., hyper-branched, linear, padlock probe), requirement of only one primer.Depending on the amplification method, product yield can be low, can give false positive result, risk of contamination resulting from either release of probes from the hybridized targets or by the removal of non-specific DNA strands from the target.Influenza A/B, Human immunodeficiency virus, Ebola virus, Zika virus, Dengue virus, Middle-East respiratory syndrome.
Quantitative Reverse Transcription PCR (RT-qPCR)Basic principle is a transcription of RNA into complementary DNA by reverse transcriptase from total RNA or mRNA, then used as template for qPCR reaction.Quantitative, it is highly specific when well-designed primers are used, high sensitivity, for high volume testing, it is amenable.Requirement of trained staff and specialized equipment, long runtime.Dengue virus, Zika virus, Chikungunya virus, Filoviruses, Hantaviruses, Influenza A, Coronavirus.
CRISPR-Cas Nucleic Acid Detection (Eg.- SHERLOCK [RPA + CRISPR], FELUDA [RPA + CRISPR] etc.)A recombinant form of the Cas9 protein with DNA endonuclease activity attaches to a single guide RNA (sgRNA), consisting of a crRNA sequence unique to the DNA target and a tracrRNA sequence interacting with the Cas9 enzyme. For the intended target, the resultant compound will specifically break double-stranded DNA. The error-prone non-homologous end joining DNA repair mechanism will repair the cleavage site, which could result in insertions or deletions that can affect gene function.Can be employed alone or with other well-known amplification techniques; due to recognition of target by both the primary amplification technique and guide RNA, this technique provides more specificity, rapid.Generally requires pre-amplification of target, may have off-target effect where cutting of wrong genes may occur due to Cas9 enzyme, risk of contamination.Ebola virus, Epstein-Barr virus, Zika virus, Dengue virus, SARS-CoV-2.
Immunochromatography (IC)Capillary action is used to carry out the immunological reaction on the chromatographic paper. Two types of particular antibodies against the antigen are used. One antibody is immobilized on chromatographic paper, while another is infiltrated into a sample pad and tagged with colloidal gold.Reliable, simple, rapid, cheap.Not sensitive, cross-reactivity checking by other tests needed.Adenovirus, Dengue virus, Puumalavirus.
MicroarrayThe concept of “nucleic acid hybridization” is the foundation of the DNA microarray. During this process, hydrogen bonds are formed between two complementary strands of DNA to create a double-stranded molecule. This makes it easier for researchers to evaluate and compare identical DNA or RNA molecules.Analysis of multiple genes, high throughput, comparison of gene expression, can categorize disease into subgroups.Result always not reproducible, expensive, false positive results, require expertization.Lymphotropic virus, Hepatitis B and C, Herpes simplex virus, Papillomavirus.
Enzyme-linked immunosorbent assay (ELISA)Specific antibodies recognize the target antigen and measure the quantity and presence of antigens binding, utilizing a substrate that changes color when the enzyme modifies it.Availability of various formats (e.g., direct, indirect, sandwich, competitive), the large spectrum of commercially available antibodies makes test design reasonably simple, suited for high-volume testing.Time-consuming, sample matrix can affect the test, different antibodies may have different target specificities, occurrence of false negative results in early infection window.Zika virus, Ebola virus, Influenza A virus, Human parechovirus, Hepatitis C, Human immunodeficiency virus.

Table 5.

Summary of diagnostic methods.

References

  1. 1. Bramhachari PV, Mohana Sheela G, Prathyusha AMVN, Madhavi M, Satish Kumar K, Reddy NNR, et al. Advanced immunotechnological methods for detection and diagnosis of viral infections: Current applications and future challenges. Dynamics of Immune Activation in Viral Diseases. 2019;5:261-275
  2. 2. Reta DH, Tessema TS, Ashenef AS, Desta AF, Labisso WL, Gizaw ST, et al. Molecular and immunological diagnostic techniques of medical viruses. International Journal of Microbiology. 2020;2020:8832728
  3. 3. Ratcliff RM, Chang G, Kok T, Sloots TP. Molecular diagnosis of medical viruses. Current Issues in Molecular Biology. 2007;9:87-102
  4. 4. Burbelo PD, Iadarola MJ, Chaturvedi A. Emerging technologies for the detection of viral infections. Future Virology. 2018;14(1):39-49. DOI: 10.2217/fvl-2018-0145
  5. 5. Influenza-seasonal. Available from: https://www.who.int/news-room [Accessed: October 20, 2023]
  6. 6. Rabies, World Health Organization. Available from: https://www.who.int/health-topics/rabies [Accessed: October 20, 2023]
  7. 7. Hepatitis, World Health Organization. Available from: https://www.who.int/health-topics/hepatitis [Accessed: October 20, 2023]
  8. 8. Alamri AM, Alkhilaiwi FA, Ullah Khan N. Era of molecular diagnostics techniques before and after the COVID-19 pandemic. Current Issues in Molecular Biology. 2022;44:4769-4789
  9. 9. Pretorius M, Venter M. Diagnosis of viral infections. Viral Infections in Children. 2017;1:151-182
  10. 10. Souf S. Recent advances in diagnostic testing for viral infections. Bioscience Horizon. 2016;9:hzw010
  11. 11. Diagnosis of respiratory syncytial virus infections, detection of RSV. Available from: http://virology-online.com/viruses/RSV6.htm [Accessed: November 20, 2023]
  12. 12. Storch GA. Diagnostic virology. Clinical Infectious Diseases. 2000;31(3):739-751
  13. 13. Hwang K-A, Ahn Ji H, Nam J-Hwan. Diagnosis of viral infections using real-time polymerase chain reaction. Journal of Bacteriology and Virology. 2018;48(1):1-13
  14. 14. Kim Y, Lovell S, Tiew KC, Mandadapu SR, Alliston KR, Battaile KP, et al. Broad-spectrum anti virals against 3C or 3C-like proteases of picorna viruses, noro viruses, and corona viruses. Journal of Virology. 2012;86(21):11754-11762
  15. 15. Dronina J, Samukaite-Bubniene U, Ramanavicius A. Advances and insights in the diagnosis of viral infections. Journal of Nanobiotechnology. 2021;19(1):348
  16. 16. Zhang Z, Liu JF, Feng T, Yao Y, Gao LH, Jiang GB. Time-resolved fluoro immunoassay as an advantageous analytical method for assessing the total concentration and environmental risk of fluoroquinolones in surface waters. Environmental Science & Technology. 2013;47:454-462
  17. 17. Alhajj M, Zubair M, Farhana A. Enzyme Linked Immunosorbent Assay. Treasure Island (FL): Stat Pearls Publishing; 2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK555922/
  18. 18. Li M, Sheng EZ, Yuan YL, Liu XF, Hua XD, Wang MH. Sensitive time-resolved fluoro immunoassay for quantitative determination of clothianidin in agricultural samples. Environmental Science and Pollution Research. 2014;21:5803-5809
  19. 19. Erkki S, Timo L, Charles BR. Time-resolved fluorescence of lanthanide probes and applications in biotechnology. CRC Critical Reviews in Analytical Chemistry. 1987;18(2):105-115
  20. 20. Tsutsumi H, Ouchi K, Ohsaki M, Yamanaka T, Kuniya Y, Takeuchi Y, et al. Immunochromatography test for rapid diagnosis of adenovirus respiratory tract infections: Comparison with virus isolation in tissue culture. Journal of Clinical Microbiology. 1999;37(6):2007-2009
  21. 21. Cinquanta L, Fontana DE, Bizzaro N. Chemiluminescent immunoassay technology: What does it change in autoantibody detection? Autoimmunity Highlights. 2017;8(1):9
  22. 22. Al-Hajjar S. Laboratory diagnosis of viral disease. In: Textbook of Clinical Pediatrics. 2012. pp. 923-928. DOI: 10.1007/978-3-642-02202-9_75
  23. 23. Oumarou Hama H, Aboudharam G, Barbieri R, Lepidi H, Drancourt M. Immuno histochemical diagnosis of human infectious diseases: A review. Diagnostic Pathology. 2022;17(1):17
  24. 24. Adone R, Sali M, Francia M, Iatarola M, Donatiello A, Fasanella A. Development of a sterne-based complement fixation test to monitor the humoral response induced by anthrax vaccines. Frontiers in Microbiology. 2016;7:19
  25. 25. Shulman NR, Barker LF. Virus-like antigen, antibody, and antigen-antibody complexes in hepatitis measured by complement fixation. Science. 1969;165(3890):304-306
  26. 26. Landry ML, Hsiung GD. DIAGNOSTIC TECHNIQUES | Isolation and identification by culture and microscopy. In: Granoff A, Webster RG, editors. Encyclopedia of Virology. 2nd ed. Connecticut, USA: Elsevier; 1999. pp. 395-403. DOI: 10.1006/rwvi.1999.0071
  27. 27. Summers MD, Volkman LE, Hsieh CH. Immuno peroxidase detection of baculo virus antigens in insect cells. The Journal of General Virology. 1978;40(3):545-557
  28. 28. Ribeiro MRC et al. Plaque reduction neutralization test (PRNT) in the congenital Zika syndrome: Positivity and associations with laboratory, clinical, and imaging characteristics. Viruses. 2020;12:1244
  29. 29. Thomas SJ, Nisalak A, Anderson KB, Libraty DH, Kalayanarooj S, Vaughn DW, et al. Dengue plaque reduction neutralization test (PRNT) in primary and secondary dengue virus infections: How alterations in assay conditions impact performance. The American Journal of Tropical Medicine and Hygiene. 2009;81(5):825-833
  30. 30. Burrell CJ, Howard CR, Murphy FA. Laboratory diagnosis of virus diseases. In: Fenner and White’s Medical Virology. 2017. pp. 135-154. DOI: 10.1016/B978-0-12-375156-0.00010-2
  31. 31. Ojha R, Nandani R, Pandey RK, Mishra A, Prajapati VK. Emerging role of circulating microRNA in the diagnosis of human infectious diseases. Journal of Cellular Physiology. 2019;234(2):1030-1043
  32. 32. Lewandowski K, Xu Y, Pullan ST, Lumley SF, Foster D, Sanderson N, et al. Metagenomic nanopore sequencing of influenza virus direct from clinical respiratory samples. Journal of Clinical Microbiology. 23 Dec 2019;58(1):e00963-19. DOI: 10.1128/JCM.00963-19
  33. 33. Mirian M, Khanahmad H, Darzi L, Salehi M, Sadeghi-Aliabadi H. Oligonucleotide aptamers: Potential novel molecules against viral hepatitis. Research in Pharmaceutical Sciences. 2017;12(2):88
  34. 34. Draz MS, Shafiee H. Applications of gold nanoparticles in virus detection. Theranostics. 2018;8(7):1985
  35. 35. Zhang H, Miller BL. Immunosensor-based label-free and multiplex detection of influenza viruses: State of the art. Biosensors & Bioelectronics. 2019;15(141):111476
  36. 36. Driskell JD, Zhu Y, Kirkwood CD, Zhao Y, Dluhy RA, Tripp RA. Rapid and sensitive detection of rotavirus molecular signatures using surface enhanced Raman spectroscopy. PLoS One. 2010;5(4):e10222
  37. 37. Saviñon-Flores F, Méndez E, López-Castaños M, Carabarin-Lima A, López-Castaños KA, González-Fuentes MA, et al. A review on SERS-based detection of human virus infections: Influenza and coronavirus. Biosensors (Basel). 2021;11(3):66
  38. 38. Duncan R, Kourout M, Grigorenka E, Fisher C, Dong M. Advances in multiplex nucleic acid diagnostics for blood-borne pathogens: Promises and pitfalls. Expert Review of Molecular Diagnostics. 2015;16:1-13
  39. 39. Cooper LA, Subbarao K. A simple restriction fragment length polymorphism-based strategy that can distinguish the internal genes of human H1N1, H3N2, and H5N1 influenza a viruses. Journal of Clinical Microbiology. 2000;38(7):2579-2583
  40. 40. Compton J. Nucleic acid sequence-based amplification. Nature. 1991;350:91-92
  41. 41. Wang H, Mo Q , Yang Z. RT-NASBA: An ideal approach for the detection of diarrhoea-related viruses. British Journal of Virology. 2015;2(3):37-42
  42. 42. Wang D, Coscoy L, Zylberberg M, Avila PC, Boushey HA, Ganem D, et al. Microarray-based detection and genotyping of viral pathogens. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(24):15687-15692
  43. 43. Mustafa MI, Makhawi AM. SHERLOCK and DETECTR: CRISPR-Cas systems as potential rapid diagnostic tools for emerging infectious diseases. Journal of Clinical Microbiology. 2021;59(3):e00745-e00720
  44. 44. Cassedy A, Parle-McDermott A, O’Kennedy R. Virus detection: A review of the current and emerging molecular and immunological methods. Frontiers in Molecular Biosciences. 2021;8:637559
  45. 45. India's Feluda COVID-19 test cheaper, faster alternative to RT-PCR, say scientists. Available from: https://economictimes.indiatimes.com/industry/healthcare/biotech/healthcare/indias-feluda-covid-19-test-cheaper-faster-alternative-to-rt-pcr-say-scientists/articleshow/78366585.cms
  46. 46. Piao J, Jiang J, Xu B, Wang X, Guan Y, Wu W, et al. Simultaneous detection and identification of enteric viruses by PCR-mass assay. PLoS One. 2012;7:e42251
  47. 47. Renuse S, Vanderboom PM, Maus AD, Kemp JV, Gurtner KM, Madugundu AK, et al. A mass spectrometry-based targeted assay for detection of SARS-CoV-2 antigen from clinical specimens. eBioMedicine. 2021;69:103465
  48. 48. Zhang J, Zhao Z, Xu M, Yang X, Liu Z. The establishment of biosensor technology based on F0F1-ATPase molecular motor for detection of rotavirus and hepatitis a virus. Biosensors Journal. 2015;4:12
  49. 49. Jung JH, Cheon DS, Liu F, Lee KB, Seo TS. A graphene oxide based immunobiosensor for pathogen detection. Angewandte Chemie (International Ed. in English). 2010;49:5708-5711
  50. 50. Baek SH, Kim MW, Park CY, Choi CS, Kailasa SK, Park JP, et al. Development of a rapid and sensitive electrochemical biosensor for detection of human norovirus via novel specific binding peptides. Biosensors & Bioelectronics. 2019;123:223-229
  51. 51. Ribeiro BV, Reis Cordeiro TA, GR EOF, Ferreira LF, Franco DL. Biosensors for the detection of respiratory viruses: A review. Talanta Open. 2020;2:100007
  52. 52. Song KK, Lee S, Ban C. Aptamers and their biological applications. Sensors. 2012;12:612-631
  53. 53. van den Kieboom CH, van der Beek SL, Mészáros T, Gyurcsányi RE, Ferwerda G, de Jonge MI. Aptasensors for viral diagnostics. Trends in Analytical Chemistry. 2015;74:58-67
  54. 54. Liu D, Zhang Z, Yin Y, Jia F, Wu Q , Tian P, et al. Development and evaluation of a novel in situ target-capture approach for aptamer selection of human noroviruses. Talanta. 2019;193:199-205
  55. 55. Sánchez-Báscones E, Parra F, Lobo-Castañón MJ. Aptamers against viruses: Selection strategies and bioanalytical applications. TrAC Trends in Analytical Chemistry. 2021;143:116349
  56. 56. Bakhtiar R. Surface plasmon resonance spectroscopy: A versatile technique in a biochemist’s toolbox. Journal of Chemical Education. 2013;90(2):203-209
  57. 57. Singh P. Surface plasmon resonance: A boon for viral diagnostics. Reference Module in Life Sciences. 2017:B978-0-12-809633-8.12245-9. DOI: 10.1016/B978-0-12-809633-8.12245-9
  58. 58. Saharan P, Khatri P, Dingolia S, Duhan JS, Gahlawat SK. Rapid detection of viruses using loop-mediated isothermal amplification (LAMP): A review. Biotechnology: Prospects and Applications. 2013;4:287-306
  59. 59. Wilkes RP, Lee PY, Tsai YL, Tsai CF, Chang HH, Chang HF, et al. An insulated isothermal PCR method on a field-deployable device for rapid and sensitive detection of canine parvovirus type 2 at points of need. Journal of Virological Methods. 2015;220:35-38
  60. 60. Piepenburg O, Williams CH, Stemple DL, Armes NA. DNA detection using recombination proteins. PLoS Biology. 2006;4(7):1115-1121
  61. 61. Euler M, Wang Y, Otto P, Tomaso H, Escudero R, Anda P, et al. Recombinase polymerase amplification assay for rapid detection of Francisella tularensis. Journal of Clinical Microbiology. 2012;50(7):2234-2238
  62. 62. Kojima K, Juma KM, Akagi S, Hayashi K, Takita T, O’Sullivan CK, et al. Solvent engineering studies on recombinase polymerase amplification. Journal of Bioscience and Bioengineering. 2020;131(2):219-224
  63. 63. Lv R, Nianhong L, Wang J, Li Y, Qi Y. Recombinase polymerase amplification for rapid detection of zoonotic pathogens: An overview. Zoonoses. 2022;2(1):6311-6331
  64. 64. Moore MD, Jaykus LA. Development of recombinase polymerase amplification assay for detection of epidemic human noroviruses. Scientific Reports. 2017;7:40244
  65. 65. Wang J, Li R, Hu L, Sun X, Wang J, Li J. Development of a quantitative fluorescence single primer isothermal amplification-based method for the detection of salmonella. International Journal of Food Microbiology. 2016;219:22-27
  66. 66. Kurn N, Chen P, Heath JD, Kopf-Sill A, Stephens KM, Wang S. Novel isothermal, linear nucleic acid amplification systems for highly multiplexed applications. Clinical Chemistry. 2005;51:1973-1981
  67. 67. Gill P, Ghaemin A. Nucleic acid isothermal amplification technologies—A review. Nucleosides, Nucleotides, and Nucleic Acids. 2008;27:224-243
  68. 68. Myrmel M, Oma V, Khatri M, Hansen HH, Stokstad M, Berg M, et al. Single primer isothermal amplification (SPIA) combined with next generation sequencing provides complete bovine coronavirus genome coverage and higher sequence depth compared to sequence-independent single primer amplification (SISPA). PLoS One. 2017;12(11):e0187780
  69. 69. Liu W, Dong D, Yang Z, Zou D, Chen Z, Yuan J, et al. Polymerase spiral reaction (PSR): A novel isothermal nucleic acid amplification method. Scientific Reports. 2015;5:12723
  70. 70. Singh TP, Jyoti KS, Sapan P, Shashi S. Polymerase spiral reaction assay for rapid and real time detection of West Nile virus from clinical samples. Frontiers in Cellular and Infection Microbiology. 2020;10:426
  71. 71. Ji J, Xu X, Wang X, et al. Novel polymerase spiral reaction assay for the visible molecular detection of porcine circovirus type 3. BMC Veterinary Research. 2019;15:322
  72. 72. Hornyák A, Bálint A, Farsang A, Balka G, Hakhverdyan M, Rasmussen TB, et al. Detection of sub genomic mRNA of feline coronavirus by real-time polymerase chain reaction based on primer-probe energy transfer (P-sg-QPCR). Journal of Virological Methods. 2012;181:155-163
  73. 73. Schaad NW, Frederick R, Shaw J, Schneider W, Hickson R, Petrillo M, et al. Advances in molecular-based diagnostics in meeting crop biosecurity and phytosanitary issues. Annual Review of Phytopathology. 2003;41:305-324
  74. 74. Rasmussen TB, Uttenthal A, Fernández J, Storgaard T. Quantitative multiplex assay for simultaneous detection and identification of Indiana and New Jersey serotypes of vesicular stomatitis virus. Journal of Clinical Microbiology. 2005;43:356-362
  75. 75. Bálint A, Tenk M, Deim Z, Rasmussen TB, Uttenthal A, Cságola A, et al. Development of primer-probe energy transfer real-time PCR for the detection and quantification of porcine circovirus type 2. Acta Veterinaria Hungarica. 2009;57(3):441-452
  76. 76. Surat P. Hybrid Capture-Based Human Papillomavirus Detection Verus PCR, Reviewed by Hanna Simmons. 2018. Available from: https://www.news-medical.net/life-sciences/Hybrid-Capture-Based-Human-Papillomavirus-Detection-Versus-PCR.aspx
  77. 77. Debaize L, Hélène Jakobczy H, Rio AG, Gandemer V, Troadec MB. Optimization of proximity ligation assay (PLA) for detection of protein interactions and fusion proteins in non-adherent cells: Application to pre-B lymphocytes. Molecular Cytogenetics. 2017;10:27
  78. 78. Liu R, He L, Hu Y, Luo Z, Zhang J. A serological aptamer-assisted proximity ligation assay for COVID-19 diagnosis and seeking neutralizing aptamers. Chemical Science. 2020;11:12157-12164
  79. 79. Barreda-García S, Miranda-Castro R, Miranda-Ordieres AJ, Lobo-Castañón MJ. Helicase-dependent isothermal amplification: A novel tool in the development of molecular-based analytical systems for rapid pathogen detection. Analytical and Bioanalytical Chemistry. 2018;410(3):679-693
  80. 80. Ciftci S, Neumann F, Abdurahman S, Appelberg KS, Mirazimi A, Nilsson M, et al. Digital rolling circle amplification–based detection of Ebola and other tropical viruses. The Journal of Molecular Diagnostics. 2020;22(2):272-283
  81. 81. Reem A, Almezgagi M, Al-Shaebi F, Al-Shehari WA, Kumal JPP. Application of rolling circle amplification (RCA) to detect the pathogens of infectious diseases. Infectious Diseases Diagnosis & Treatment. 2020;4:158
  82. 82. Zhu X, Wang Y, Chen J, Zhang X, Shi H, Shi D, et al. Development of TaqMan real-time reverse transcription-polymerase chain reaction for the detection and quantitation of porcine kobuvirus. Journal of Virological Methods. 2016;234:132-136
  83. 83. Vilibic-Cavlek T, Savic V, Ferenc T, Mrzljak A, Barbic L, Bogdanic M, et al. Lymphocytic choriomeningitis—emerging trends of a neglected virus: A narrative review. Tropical Medicine and Infectious Disease. 2021;6:88
  84. 84. Okitsu S, Khamrin P, Thongprachum A, Nishimura S, Kalesaran A, Takanashi S, et al. Detection and molecular characterization of human Cosa virus in a paediatric patient with acute gastroenteritis, Japan. Infection, Genetics and Evolution. 2014;28:125-129
  85. 85. Grubman MJ, Barry BB. Foot and mouth disease. Clinical Microbiology Reviews. 2004;17(2):465-493
  86. 86. Hasan S, Jamdar SF, Alalowi M, Al A, Beaiji SM. Dengue virus: A global human threat: Review of literature. Journal of International Society of Preventive & Community Dentistry. 2016;6:1-6
  87. 87. Broadhurst MJ, Brooks GTJ, Pollock NR. Diagnosis of Ebola virus disease: Past, present, and future. Clinical Microbiology Reviews. 2016;29(4):773-793
  88. 88. Wells AI, Coyne CB. Enteroviruses: A gut-wrenching game of entry, detection, and evasion. Viruses. 2019;11(5):460
  89. 89. Liang TJ. Hepatitis B: The virus and disease. Hepatology. 2009;49(5):13-21
  90. 90. Guan TP, Teng JLL, Yeong KY, You ZQ , Liu H, Wong SSY, et al. Metagenomic analysis of Sichuan takin fecal sample viromes reveals novel enterovirus and astrovirus. Virology. 2018;521:77-91
  91. 91. Pérot P, Lecuit M, Eloit M. Astrovirus diagnostics. Viruses. 2017;9:1
  92. 92. Branson BM. The future of HIV testing. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2010;55:S102-S105
  93. 93. van Leeuwen M, Williams MM, Koraka P, Simon JH, Smits SL, Osterhaus AD. Human picobirna viruses identified by molecular screening of diarrhea samples. Journal of Clinical Microbiology. 2010;48(5):1787-1794
  94. 94. Jacobs SE, Lamson DM, St George K, Walsh TJ. Human rhinoviruses. Clinical Microbiology Reviews. 2013;26(1):135-162
  95. 95. van Maarseveen NM, Wessels E, de Brouwer CS, Vossen AC, Claas EC. Diagnosis of viral gastroenteritis by simultaneous detection of adenovirus group F, astrovirus, rotavirus group a, norovirus genogroups I and II, and sapovirus in two internally controlled multiplex real-time PCR assays. Journal of Clinical Virology. 2010;49:205-210
  96. 96. Kumar N, Malik YS, Kumar S, Sharma K, Sircar S, Saurabh S, et al. Peptide-recombinant VP6 protein based enzyme immunoassay for the detection of group a rotaviruses in multiple host species. PLoS One. 2016;11:e0159027
  97. 97. Ye X, Xu J, Lu L, Li X, Fang X, Kong J. Equipment-free nucleic acid extraction and amplification on a simple paper disc for point-of-care diagnosis of rotavirus A. Analytica Chimica Acta. 2018;1018:78-85
  98. 98. Varela MF, Monteiro S, Rivadulla E, Santos R, Romalde JL. Development of a novel digital RT-PCR method for detection of human sapovirus in different matrices. Journal of Virological Methods. 2018;254:21-24
  99. 99. Dziąbowska K, Czaczyk E, Nidzworski D. Detection methods of human and animal influenza virus—Current trends. Biosensors. 2018;8(4):94
  100. 100. Solomon T, Thao LT, Dung NM, Kneen R, Hung NT, Nisalak A, et al. Rapid diagnosis of Japanese encephalitis by using an immunoglobulin M dot enzyme immunoassay. Journal of Clinical Microbiology. 1998;36(7):2030-2034
  101. 101. Mansfield KL, Hernández-Triana LM, Banyard AC, Fooks AR, Johnson N. Japanese encephalitis virus infection, diagnosis and control in domestic animal. Veterinary Microbiology. 2017;201:85-92
  102. 102. Fooks AR, Cliquet F, Finke S, Freuling C, Hemachudha T, Mani RS, et al. Rabies. Nature Reviews. Disease Primers. 2017;3:17091
  103. 103. Lelli D, Beato MS, Cavicchio L, Lavazza A, Chiapponi C, Leopardi S, et al. First identification of mammalian orthoreovirus type 3 in diarrheic pigs in Europe. Virology Journal. 2016;13:139
  104. 104. Zhang W, Kataoka M, Doan YH, Oi T, Furuya T, Oba M, et al. Isolation and characterization of mammalian orthoreovirus type 3 from a fecal sample from a wild boar in Japan. Archives of Virology. 2021;166(6):1671-1680
  105. 105. Nasheri N, Petronella N, Ronholm J, Bidawid S, Corneau N. Characterization of the genomic diversity of norovirus in linked patients using a metagenomic deep sequencing approach. Frontiers in Microbiology. 2017;8:73
  106. 106. Branche AR, Falsey AR. Parainfluenza virus infection. Seminars in Respiratory and Critical Care Medicine. 2016;37:538-554
  107. 107. Vaidya SR, Hamde VS, Kumbhar NS, Walimbe AM. Utility of neutralization test for laboratory diagnosis of suspected mumps. Microbiology and Immunology. 2018;62(4):243-247
  108. 108. Okafuji T, Yoshida N, Fujino M, Motegi Y, Ihara T, Ota Y, et al. Rapid diagnostic method for detection of mumps virus genome by loop-mediated isothermal amplification. Journal of Clinical Microbiology. 2005;43(4):1625-1631
  109. 109. Mehndiratta MM, Mehndiratta P, Pande R. Poliomyelitis: Historical facts, epidemiology, and current challenges in eradication. The Neurohospitalist. 2014;4(4):223-229
  110. 110. Eiland LS. Respiratory syncytial virus: Diagnosis, treatment and prevention. Journal of Pediatric Pharmacology and Therapeutics. 2009;14:75-85
  111. 111. Rota PA, Moss WJ, Takeda M, de Swart RL, Thompson KM, Goodson JL. Measles. Primer. 2016;2:1-16
  112. 112. Ayouni S, Estienney M, Hammami S, Guediche MN, Pothier P, Aouni M, et al. Cosavirus, salivirus and bufavirus in diarrheal tunisian infants. PLoS One. 2016;11:e0162255
  113. 113. Honkavuori KS, Briese T, Krauss S, Sanchez MD, Jain K, Hutchison SK, et al. Novel coronavirus and astrovirus in Delaware Bay shorebirds. PLoS One. 2014;9(4):e93395
  114. 114. Kevadiya BD, Machhi J, Herskovitz J, Oleynikov MD, Blomberg WR, Bajwa N, et al. Diagnostics for SARS-CoV-2 infections. Nature Materials. 2021;20(5):593-605
  115. 115. Liu X, Zhou Y, Yang F, Liu P, Cai Y, Huang J, et al. Rapid and sensitive detection of porcine torovirus by a reverse transcription loop-mediated isothermal amplification assay (RT-LAMP). Journal of Virological Methods. 2016;228:103-107
  116. 116. Colpitts TM, Conway MJ, Montgomery RR, Fikriga E. West Nile virus: Biology, transmission, and human infection. Clinical Microbiology Reviews. 2012;25(4):635-648
  117. 117. Dela Cruz CS, Pasnick S, Gross JE, Keller J, Carlos GW, Cao B, et al. Adenovirus infection and outbreaks: What you need to know. American Journal of Respiratory and Critical Care Medicine. 2019;199:13-14
  118. 118. Henke-Gendo C, Ganzenmueller T, Kluba J, Harste G, Raggub L, et al. Improved quantitative PCR protocols for adenovirus and CMV with an internal inhibition control system and automated nucleic acid isolation. Journal of Medical Virology. 2012;84(6):890-896
  119. 119. Pabbaraju K, Wong S, Fox JD. Detection of adenoviruses. In: Stephenson JR, Warnes A, editors. Diagnostic Virology Protocols. Totowa, NJ: Humana Press; 2010. pp. 1-15
  120. 120. Fugl A, Andersen CL. Epstein-Barr virus and its association with disease - a review of relevance to general practice. BMC Family Practice. 2019;20:62
  121. 121. Koelle DM, Corey L. Recent progress in herpes simplex virus immunobiology and vaccine research. Clinical Microbiology Reviews. 2003;16(1):96-113
  122. 122. Pfeffer M, Meyer H. Poxvirus diagnostics. In: Poxviruses. Birkhäuser; 2007. pp. 355-373
  123. 123. Gershon AA, Breuer J, Cohen JI, Cohrs RJ, Gershon MD, Gilden D, et al. Varicella zoster virus infection. Nature Reviews Disease Primers. 2 Jul 2015;1:15016. DOI: 10.1038/nrdp.2015.16
  124. 124. Kostoulas P, Eusebi P, Hartnack S. Diagnostic accuracy estimates for COVID-19 real-time polymerase chain reaction and lateral flow immunoassay tests with Bayesian latent-class models. American Journal of Epidemiology. 2021;190(8):1689-1695
  125. 125. Mat Jusoh TNA, Shueb RH. Performance evaluation of commercial dengue diagnostic tests for early detection of dengue in clinical samples. Journal of Tropical Medicine. 2017;2017:4687182
  126. 126. Saboor Soomro R, Shah IA, Saboor A, Bhutto AUB, Memon S. Sensitivity and specificity of hepatitis B virus screening via rapid immunoassay chromatographic test. Cureus. 2021;13(1):e12909
  127. 127. Owens DK, Holodniy M, McDonald TW, Scott J, Sonnad S. A meta-analytic evaluation of the polymerase chain reaction for the diagnosis of HIV infection in infants. Journal of the American Medical Association. 1996;275(17):1342-1348
  128. 128. Solomon T, Dung NM, Kneen R, Gainsborough M, Vaughn DW, Khanh VT. Japanese encephalitis. Journal of Neurology, Neurosurgery, and Psychiatry. 2000;68(4):405-415
  129. 129. Rasolonjatovo FS, Guis H, Rajeev M, Dacheux L, Arivony Nomenjanahary L, Razafitrimo G, et al. Enabling animal rabies diagnostic in low-access areas: Sensitivity and specificity of a molecular diagnostic test from cerebral tissue dried on filter paper. PLoS Neglected Tropical Diseases. 2020;14(3):e0008116
  130. 130. Zubach V, Severini A, Hiebert J. Development of a rapid, internally controlled, two targets, real-time RT-PCR for detection of measles virus. Journal of Virological Methods. 2022;2021(299):114349
  131. 131. Park SY, Kim JY, Kim JA, Kwon JS, Kim SM, Jeon NY, et al. Diagnostic usefulness of varicella-zoster virus real-time polymerase chain reaction analysis of DNA in saliva and plasma specimens from patients with herpes zoster. The Journal of Infectious Diseases. 2017;217(1):51-57
  132. 132. Hadfield TL, Turell M, Dempsey MP, David J, Park EJ. Detection of West Nile virus in mosquitoes by RT-PCR. Molecular and Cellular Probes. 2001;15(3):147-150
  133. 133. Soheili M, Keyvani H, Soheili M, Nasseri S. Human papilloma virus: A review study of epidemiology, carcinogenesis, diagnostic methods, and treatment of all HPV related cancers. Medical Journal of the Islamic Republic of Iran. 2021;35:65

Written By

Biswajyoti Borkakoty, Aniruddha Jakharia, Pratibha Singh and Siraj Ahmed Khan

Submitted: 02 August 2023 Reviewed: 18 September 2023 Published: 06 March 2024